Humanized mouse

Last updated

A humanized mouse is a genetically modified mouse that has functioning human genes, cells, tissues and/or organs. [1] Humanized mice are commonly used as small animal models in biological and medical research for human therapeutics. [2]

Contents

A humanized mouse or a humanized mouse model is one that has been xenotransplanted with human cells and/or engineered to express human gene products, so as to be utilized for gaining relevant insights in the in vivo context for understanding of human-specific physiology and pathologies. [3] A lot of our knowledge about several human biological processes has been obtained from studying animal models like rodents and non-human primates. In particular, small animals such as mice are advantageous in such studies owing to their small size, brief reproductive cycle, easy handling and due to the genomic and physiological similarities with humans; moreover, these animals can also be genetically modified easily. Nevertheless, there are several incongruencies of these animal systems with those of humans, especially with regard to the components of the immune system. To overcome these limitations and to realize the full potential of animal models to enable researchers to get a clear picture of the nature and pathogenesis of immune responses mounted against human-specific pathogens, humanized mouse models have been developed. Such mouse models have also become an integral aspect of preclinical biomedical research. [4]

History

The discovery of the athymic mouse, commonly known as the nude mouse, and that of the SCID mouse were major events that paved the way for humanized mice models. The first such mouse model was derived by backcrossing C57BL/Ka and BALB/c mice, featuring a loss of function mutation in the PRKDC gene. The PRKDC gene product is necessary for resolving breaks in DNA strands during the development of T cells and B cells. A mutation in the Foxn1 gene on chromosome 11 resulted in impaired thymus development, leading to a deficiency in mature T lymphocytes. Dysfunctional PRKDC gene leads to impaired development of T and B lymphocytes which gives rise to severe combined immunodeficiency (SCID). In spite of the efforts in developing this mouse model, poor engraftment of human hematopoietic stem cells (HSCs) was a major limitation that called for further advancement in the development humanized mouse models. [5] Nude mice were the earliest immunodeficient mouse model. These mice primarily produced IgM and had minimal or no IgA. As a result, they did not exhibit a rejection response to allogeneic tissue. Commonly utilized strains included BALB/c-nu, Swiss-nu, NC-nu, and NIH-nu, which were extensively employed in the research of immune diseases and tumors. However, due to the retention of B cells and NK cells, they were unable to fully support engraftment of human immune cells, thus making them unsuitable as an ideal humanized mouse model.

The next big step in the development of humanized mice models came with transfer of the scid mutation to a non-obese diabetic mouse. This resulted in the creation of the NOD-scid mice which lacked T cells, B cells, and NK cells. This mouse model permitted for a slightly higher level of human cell reconstitution. Nevertheless, a major breakthrough in this field came with the introduction of the mutant IL-2 receptor (IL2rg) gene in the NOD-scid model. This accounted for the creation of the NOD-scid-γcnull mice (NCG, NSG or NOG) models which were found to have defective signaling of interleukins IL-2, IL-4, IL-7, IL-9, IL-15 and IL-21. Researchers evolved this NSG model by knocking out the RAG1 and RAG2 genes (recombination activation genes), resulting into the RAGnull version of the NSG model that was devoid of major cells of the immune system including the natural killer cells, B lymphocytes and T lymphocytes, macrophages and dendritic cells, causing the greatest immunodeficiency in mice models so far. The limitation with this model was that it lacked the human leukocyte antigen. In accordance to this limitation, the human T cells when engrafted in the mice, failed to recognize human antigen-presenting cells, which consequated in defective immunoglobulin class switching and improper organization of the secondary lymphoid tissue. [6]

To circumvent this limitation, the next development came with the introduction of transgenes encoding for HLA I and HLA II in the NSG RAGnull model that enabled buildout of human T-lymphocyte repertoires as well as the respective immune responses. [7] Mice with such human genes are technically human-animal hybrids.

Types

Engrafting an immunodeficient mouse with functional human cells can be achieved by intravenous injections of human cells and tissue into the mouse, and/or creating a genetically modified mouse from human genes. These models have been instrumental in studying human diseases, immune responses, and therapeutic interventions. This section highlights the various humanized mice models developed using the different methods.

Hu-PBL-scid model

The human peripheral blood lymphocyte-severe combined immunodeficiency mouse model has been employed in a diverse array of research, encompassing investigations into Epstein-Barr virus (EBV)-associated lymphoproliferative disease, toxoplasmosis, human immunodeficiency virus (HIV) infection, and autoimmune diseases. [8] These studies have highlighted the effectiveness of the hu-PBL-SCID mouse model in examining various facets of human diseases, including pathogenesis, immune responses, and therapeutic interventions. Furthermore, the model has been utilized to explore genetic and molecular factors linked to neuropsychiatric disorders such as schizophrenia, offering valuable insights into the pathophysiology and potential therapeutic targets for these conditions. [9] This model is developed by intravenously injecting human PBMCs into immunodeficient mice. The peripheral blood mononuclear cells to be engrafted into the model are obtained from consented adult donors. The advantages associated with this method are that it is comparatively an easy technique, the model takes relatively less time to get established and that the model exhibits functional memory T cells. [10] It is particularly very effective for modelling graft vs. host disease. [7] The model lacks engraftment of B lymphocytes and myeloid cells. Other limitations with this model are that it is suitable for use only in short-term experiments (<3 months) and the possibility that the model itself might develop graft vs. host disease. [7]

Hu-SRC-scid model

The humanized severe combined immunodeficiency (SCID) mouse model, also known as the hu-SRC-scid model, has been extensively utilized in various research areas, including immunology, infectious diseases, cancer, and drug development. This model has been instrumental in studying the human immune response to xenogeneic and allogeneic decellularized biomaterials, providing valuable insights into the biocompatibility and gene expression regulation of these materials. [11] Hu-SRC-scid mice are developed by engrafting CD34+ human hematopoietic stem cells into immunodeficient mice. The cells are obtained from human fetal liver, bone marrow or from blood derived from the umbilical cord, [12] and engrafted via intravenous injection. The advantages of this model are that it offers multilineage development of hematopoietic cells, generation of a naïve immune system, and if engraftment is carried out by intrahepatic injection of newborn mice within 72 hours of birth, it can lead to enhanced human cell reconstitution. Nevertheless, limitations associated with the model are that it takes a minimum of 10 weeks for cell differentiation to occur, it harbors low levels of human RBCs, polymorphonuclear leukocytes, and megakaryocytes. [7]

BLT (bone marrow/liver/thymus) model

The BLT model is constituted with human HSCs, bone marrow, liver, and thymus. The engraftment is carried out by implantation of liver and thymus under the kidney capsule and by transplantation of HSCs obtained from fetal liver. The BLT model has a complete and totally functional human immune system with HLA-restricted T lymphocytes. The model also comprises a mucosal system that is similar to that of humans. Moreover, among all models the BLT model has the highest level of human cell reconstitution. [13]

However, since it requires surgical implantation, this model is the most difficult and time-consuming to develop. Other drawbacks associated with the model are that it portrays weak immune responses to xenobiotics, sub-optimal class switching and may develop GvHD. [7]

Transplanted human organoids

Bio- and electrical engineers have shown that human cerebral organoids transplanted into mice functionally integrate with their visual cortex. [14] [15] Such models may raise similar ethical issues as organoid-based humanization of other animals.

Mouse-human hybrid

A mouse-human hybrid is a genetically modified mouse whose genome has both mouse and human genes, thus being a murine form of a human-animal hybrid. For example, genetically modified mice may be born with human leukocyte antigen genes in order to provide a more realistic environment when introducing human white blood cells into them in order to study immune system responses. [7] One such application is the identification of hepatitis C virus (HCV) peptides that bind to HLA, and that can be recognized by the human immune system, thereby potentially being targets for future vaccines against HCV. [16]

Established models for human diseases

Several mechanisms underlying human maladies are not fully understood. Utilization of humanized mice models in this context allows researchers to determine and unravel important factors that bring about the development of several human diseases and disorders falling under the categories of infectious disease, cancer, autoimmunity, and GvHD.

Infectious diseases

Among the human-specific infectious pathogens studied on humanized mice models, the human immunodeficiency virus has been successfully studied. [7] Besides this, humanized models for studying Ebola virus, [17] Hepatitis B, [18] Hepatitis C, [19] Kaposi's sarcoma-associated herpesvirus, [20] Leishmania major, [21] malaria, [22] and tuberculosis [23] have been reported by various studies.

NOD/scid mice models for dengue virus [24] and varicella-zoster virus, [25] and a Rag2null𝛾cnull model for studying influenza virus [26] have also been developed.

Cancers

On the basis of the type of human cells/tissues that have been used for engraftment, humanized mouse models for cancer can be classified as patient-derived xenografts or cell line-derived xenografts. [27] PDX models are considered to retain the parental malignancy characteristics at a greater extent and hence these are regarded as the more powerful tool for evaluating the effect of anticancer drugs in pre-clinical studies. [27] [28] Humanized mouse models for studying cancers of various organs have been designed. A mouse model for the study of breast cancer has been generated by the intrahepatic engraftment of SK-BR-3 cells in NSG mice. [29] Similarly, NSG mice intravenously engrafted with patient-derived AML cells, [30] and those engrafted (via subcutaneous, intravenous or intra-pancreatic injections) with patient-derived pancreatic cancer tumors [31] have also been developed for the study of leukemia and pancreatic cancer respectively. Several other humanized rodent models for the study of cancer and cancer immunotherapy have also been reported. [32]

Autoimmune diseases

Problems posed by the differences in the human and rodent immune systems have been overcome using a few strategies, so as to enable researchers to study autoimmune disorders using humanized models. As a result, the use of humanized mouse models has extended to various areas of immunology and disease research. For instance, humanized mice have been utilized to study human-tropic pathogens, liver cancer models, and the comparison of mouse models to human diseases NSG mice engrafted with PBMCs and administered with myelin antigens in Freund's adjuvant, and antigen-pulsed autologous dendritic cells have been used to study multiple sclerosis. [33] Similarly, NSG mice engrafted with hematopoietic stem cells and administered with pristane have been used for studying lupus erythematosus. [34] Furthermore, NOG mice engrafted with PBMCs has been used to study mechanisms of allografts rejection in vivo. [35] The development of humanized mouse models has significantly advanced the study of autoimmune disorders and various areas of immunology and disease research. These models have provided a platform for investigating human diseases, immune responses, and therapeutic interventions, bridging the gap between human and rodent immune systems and offering valuable insights into disease pathogenesis and potential therapeutic strategies.

See also

Related Research Articles

<span class="mw-page-title-main">Cytotoxic T cell</span> T cell that kills infected, damaged or cancerous cells

A cytotoxic T cell (also known as TC, cytotoxic T lymphocyte, CTL, T-killer cell, cytolytic T cell, CD8+ T-cell or killer T cell) is a T lymphocyte (a type of white blood cell) that kills cancer cells, cells that are infected by intracellular pathogens (such as viruses or bacteria), or cells that are damaged in other ways.

<span class="mw-page-title-main">Severe combined immunodeficiency</span> Genetic disorder leading to severe impairment of the immune system

Severe combined immunodeficiency (SCID), also known as Swiss-type agammaglobulinemia, is a rare genetic disorder characterized by the disturbed development of functional T cells and B cells caused by numerous genetic mutations that result in differing clinical presentations. SCID involves defective antibody response due to either direct involvement with B lymphocytes or through improper B lymphocyte activation due to non-functional T-helper cells. Consequently, both "arms" of the adaptive immune system are impaired due to a defect in one of several possible genes. SCID is the most severe form of primary immunodeficiencies, and there are now at least nine different known genes in which mutations lead to a form of SCID. It is also known as the bubble boy disease and bubble baby disease because its victims are extremely vulnerable to infectious diseases and some of them, such as David Vetter, have become famous for living in a sterile environment. SCID is the result of an immune system so highly compromised that it is considered almost absent.

<span class="mw-page-title-main">Adenosine deaminase deficiency</span> Medical condition

Adenosine deaminase deficiency is a metabolic disorder that causes immunodeficiency. It is caused by mutations in the ADA gene. It accounts for about 10–20% of all cases of autosomal recessive forms of severe combined immunodeficiency (SCID) after excluding disorders related to inbreeding.

Mouse mammary tumor virus (MMTV) is a milk-transmitted retrovirus like the HTL viruses, HI viruses, and BLV. It belongs to the genus Betaretrovirus. MMTV was formerly known as Bittner virus, and previously the "milk factor", referring to the extra-chromosomal vertical transmission of murine breast cancer by adoptive nursing, demonstrated in 1936, by John Joseph Bittner while working at the Jackson Laboratory in Bar Harbor, Maine. Bittner established the theory that a cancerous agent, or "milk factor", could be transmitted by cancerous mothers to young mice from a virus in their mother's milk. The majority of mammary tumors in mice are caused by mouse mammary tumor virus.

<span class="mw-page-title-main">Omenn syndrome</span> Medical condition

Omenn syndrome is an autosomal recessive severe combined immunodeficiency. It is associated with hypomorphic missense mutations in immunologically relevant genes of T-cells such as recombination activating genes, Interleukin-7 receptor-α (IL7Rα), DCLRE1C-Artemis, RMRP-CHH, DNA-Ligase IV, common gamma chain, WHN-FOXN1, ZAP-70 and complete DiGeorge syndrome. It is fatal without treatment.

<span class="mw-page-title-main">X-linked severe combined immunodeficiency</span> Medical condition

X-linked severe combined immunodeficiency (X-SCID) is an immunodeficiency disorder in which the body produces very few T cells and NK cells.

Primary immunodeficiencies are disorders in which part of the body's immune system is missing or does not function normally. To be considered a primary immunodeficiency (PID), the immune deficiency must be inborn, not caused by secondary factors such as other disease, drug treatment, or environmental exposure to toxins. Most primary immunodeficiencies are genetic disorders; the majority are diagnosed in children under the age of one, although milder forms may not be recognized until adulthood. While there are over 430 recognized inborn errors of immunity (IEIs) as of 2019, the vast majority of which are PIDs, most are very rare. About 1 in 500 people in the United States are born with a primary immunodeficiency. Immune deficiencies can result in persistent or recurring infections, auto-inflammatory disorders, tumors, and disorders of various organs. There are currently limited treatments available for these conditions; most are specific to a particular type of PID. Research is currently evaluating the use of stem cell transplants (HSCT) and experimental gene therapies as avenues for treatment in limited subsets of PIDs.

<span class="mw-page-title-main">WHIM syndrome</span> Medical condition

WHIM syndrome is a rare congenital immunodeficiency disorder characterized by chronic noncyclic neutropenia.

<span class="mw-page-title-main">C-C chemokine receptor type 7</span> Protein-coding gene in the species Homo sapiens

C-C chemokine receptor type 7 is a protein that in humans is encoded by the CCR7 gene. Two ligands have been identified for this receptor: the chemokines ligand 19 (CCL19/ELC) and ligand 21 (CCL21). The ligands have similar affinity for the receptor, though CCL19 has been shown to induce internalisation of CCR7 and desensitisation of the cell to CCL19/CCL21 signals. CCR7 is a transmembrane protein with 7 transmembrane domains, which is coupled with heterotrimeric G proteins, which transduce the signal downstream through various signalling cascades. The main function of the receptor is to guide immune cells to immune organs by detecting specific chemokines, which these tissues secrete.

<span class="mw-page-title-main">GPR15</span> Protein-coding gene in the species Homo sapiens

G protein-coupled receptor 15 is a protein that in humans is encoded by the GPR15 gene.

The severe combined immunodeficiency (SCID) is a severe immunodeficiency genetic disorder that is characterized by the complete inability of the adaptive immune system to mount, coordinate, and sustain an appropriate immune response, usually due to absent or atypical T and B lymphocytes. In humans, SCID is colloquially known as "bubble boy" disease, as victims may require complete clinical isolation to prevent lethal infection from environmental microbes.

<span class="mw-page-title-main">CD226</span> Protein-coding gene in the species Homo sapiens

CD226, PTA1 or DNAM-1 is a ~65 kDa immunoglobulin-like transmembrane glycoprotein expressed on the surface of natural killer cells, NK T cell, B cells, dendritic cells, hematopoietic precursor cells, platelets, monocytes and T cells.

<span class="mw-page-title-main">CD200</span> Protein-coding gene in the species Homo sapiens

OX-2 membrane glycoprotein, also named CD200 is a human protein encoded by the CD200 gene. CD200 gene is in human located on chromosome 3 in proximity to genes encoding other B7 proteins CD80/CD86. In mice CD200 gene is on chromosome 16.

<span class="mw-page-title-main">BCL11B</span> Protein-coding gene in the species Homo sapiens

B-cell lymphoma/leukemia 11B is a protein that in humans is encoded by the BCL11B gene.

A NOG (NOD/Shi-scid/IL-2Rγnull) mouse is a new generation of severely immunodeficient mouse, developed by Central Institute for Experimental Animals (CIEA) in 2000. The NOG mouse accepts heterologous cells much more easily compared with any other type of immunodeficient rodent models, such as nude mouse and NOD/scid mouse. Thus, the mouse can be the best model as a highly efficient recipient of human cells to engraft, proliferate and differentiate. This unique feature offers a great opportunity for enhancing therapy researches of cancer, leukemia, visceral diseases, AIDS, and other human diseases. It also provides applications for cancer, infection, regeneration, and hematology researches.

<span class="mw-page-title-main">Genetically modified mouse</span>

A genetically modified mouse or genetically engineered mouse model (GEMM) is a mouse that has had its genome altered through the use of genetic engineering techniques. Genetically modified mice are commonly used for research or as animal models of human diseases and are also used for research on genes. Together with patient-derived xenografts (PDXs), GEMMs are the most common in vivo models in cancer research. Both approaches are considered complementary and may be used to recapitulate different aspects of disease. GEMMs are also of great interest for drug development, as they facilitate target validation and the study of response, resistance, toxicity and pharmacodynamics.

The NSG mouse is a brand of immunodeficient laboratory mice, developed and marketed by Jackson Laboratory, which carries the strain NOD.Cg-Prkdcscid Il2rgtm1Wjl/SzJ. NSG branded mice are among the most immunodeficient described to date. NSG branded mice lack mature T cells, B cells, and natural killer (NK) cells. NSG branded mice are also deficient in multiple cytokine signaling pathways, and they have many defects in innate immunity. The compound immunodeficiencies in NSG branded mice permit the engraftment of a wide range of primary human cells, and enable sophisticated modeling of many areas of human biology and disease. NSG branded mice were developed in the laboratory of Dr. Leonard Shultz at Jackson Laboratory, which owns the NSG trade mark.

Patient derived xenografts (PDX) are models of cancer where the tissue or cells from a patient's tumor are implanted into an immunodeficient or humanized mouse. It is a form of xenotransplantation. PDX models are used to create an environment that allows for the continued growth of cancer after its removal from a patient. In this way, tumor growth can be monitored in the laboratory, including in response to potential therapeutic options. Cohorts of PDX models can be used to determine the therapeutic efficiency of a therapy against particular types of cancer, or a PDX model from a specific patient can be tested against a range of therapies in a 'personalized oncology' approach.

Mice with severe combined immunodeficiency (SCIDs) are often used in the research of human disease. Human immune cells are used to develop human lymphoid organs within these immunodeficient mice, and many different types of SCID mouse models have been developed. These mice allow researchers to study the human immune system and human disease in a small animal model.

<span class="mw-page-title-main">Mouse Models of Human Cancer database</span>

The laboratory mouse has been instrumental in investigating the genetics of human disease, including cancer, for over 110 years. The laboratory mouse has physiology and genetic characteristics very similar to humans providing powerful models for investigation of the genetic characteristics of disease.

References

  1. Chuprin J, Buettner H, Seedhom MO, Greiner DL, Keck JG, Ishikawa F, et al. (March 2023). "Humanized mouse models for immuno-oncology research". Nature Reviews. Clinical Oncology. 20 (3): 192–206. doi:10.1038/s41571-022-00721-2. PMC   10593256 . PMID   36635480.
  2. Brehm MA, Wiles MV, Greiner DL, Shultz LD (August 2014). "Generation of improved humanized mouse models for human infectious diseases". Journal of Immunological Methods. 410: 3–17. doi:10.1016/j.jim.2014.02.011. PMC   4155027 . PMID   24607601.
  3. Stripecke R, Münz C, Schuringa JJ, Bissig KD, Soper B, Meeham T, et al. (July 2020). "Innovations, challenges, and minimal information for standardization of humanized mice". EMBO Molecular Medicine. 12 (7): e8662. doi:10.15252/emmm.201708662. PMC   7338801 . PMID   32578942.
  4. Walsh NC, Kenney LL, Jangalwe S, Aryee KE, Greiner DL, Brehm MA, Shultz LD (January 2017). "Humanized Mouse Models of Clinical Disease". Annual Review of Pathology. 12 (1): 187–215. doi:10.1146/annurev-pathol-052016-100332. PMC   5280554 . PMID   27959627.
  5. Ito R, Takahashi T, Katano I, Ito M (May 2012). "Current advances in humanized mouse models". Cellular & Molecular Immunology. 9 (3): 208–14. doi:10.1038/cmi.2012.2. PMC   4012844 . PMID   22327211.
  6. Bosma GC, Custer RP, Bosma MJ (February 1983). "A severe combined immunodeficiency mutation in the mouse". Nature. 301 (5900): 527–30. Bibcode:1983Natur.301..527B. doi:10.1038/301527a0. PMID   6823332. S2CID   4267981.
  7. 1 2 3 4 5 6 7 Yong KS, Her Z, Chen Q (August 2018). "Humanized Mice as Unique Tools for Human-Specific Studies". Archivum Immunologiae et Therapiae Experimentalis. 66 (4): 245–266. doi:10.1007/s00005-018-0506-x. PMC   6061174 . PMID   29411049.
  8. Ahmed EH, Baiocchi RA (2016-03-31). "Murine Models of Epstein-Barr Virus-Associated Lymphomagenesis". ILAR Journal. 57 (1): 55–62. doi:10.1093/ilar/ilv074. PMC   6302257 . PMID   27034395.
  9. Nomura J, Takumi T (2012). "Animal models of psychiatric disorders that reflect human copy number variation". Neural Plasticity. 2012: 589524. doi: 10.1155/2012/589524 . PMC   3414062 . PMID   22900207.
  10. Tary-Lehmann M, Saxon A, Lehmann PV (November 1995). "The human immune system in hu-PBL-SCID mice". Immunology Today. 16 (11): 529–533. doi:10.1016/0167-5699(95)80046-8. PMID   7495490.
  11. Wang RM, Johnson TD, He J, Rong Z, Wong M, Nigam V, et al. (June 2017). "Humanized mouse model for assessing the human immune response to xenogeneic and allogeneic decellularized biomaterials". Biomaterials. 129: 98–110. doi:10.1016/j.biomaterials.2017.03.016. PMC   5434867 . PMID   28334641.
  12. Pearson T, Greiner DL, Shultz LD (May 2008). "Creation of "humanized" mice to study human immunity". Current Protocols in Immunology. Chapter 15 (1): 15.21.1–15.21.21. doi:10.1002/0471142735.im1521s81. PMC   3023233 . PMID   18491294.
  13. Karpel ME, Boutwell CL, Allen TM (August 2015). "BLT humanized mice as a small animal model of HIV infection". Current Opinion in Virology. Animal models for viral diseases / Oncolytic viruses. 13: 75–80. doi:10.1016/j.coviro.2015.05.002. PMC   4550544 . PMID   26083316.
  14. Firtina N (3 January 2023). "In a first, human brain organoids placed in the mouse cortex react to visual stimuli". Interesting Engineering . Retrieved 17 January 2023.
  15. Wilson MN, Thunemann M, Liu X, Lu Y, Puppo F, Adams JW, et al. (December 2022). "Multimodal monitoring of human cortical organoids implanted in mice reveal functional connection with visual cortex". Nature Communications. 13 (1): 7945. Bibcode:2022NatCo..13.7945W. doi: 10.1038/s41467-022-35536-3 . PMC   9792589 . PMID   36572698.
  16. "Mouse strain C57BL/6-Mcph1Tg(HLA-A2.1)1Enge". The Jackson Laboratory. Retrieved 2023-01-06.
  17. Lüdtke A, Oestereich L, Ruibal P, Wurr S, Pallasch E, Bockholt S, et al. (April 2015). "Ebola virus disease in mice with transplanted human hematopoietic stem cells". Journal of Virology. 89 (8): 4700–4. doi:10.1128/JVI.03546-14. PMC   4442348 . PMID   25673711.
  18. Bility MT, Cheng L, Zhang Z, Luan Y, Li F, Chi L, et al. (March 2014). "Hepatitis B virus infection and immunopathogenesis in a humanized mouse model: induction of human-specific liver fibrosis and M2-like macrophages". PLOS Pathogens. 10 (3): e1004032. doi: 10.1371/journal.ppat.1004032 . PMC   3961374 . PMID   24651854.
  19. Bility MT, Zhang L, Washburn ML, Curtis TA, Kovalev GI, Su L (September 2012). "Generation of a humanized mouse model with both human immune system and liver cells to model hepatitis C virus infection and liver immunopathogenesis". Nature Protocols. 7 (9): 1608–17. doi:10.1038/nprot.2012.083. PMC   3979325 . PMID   22899330.
  20. Wang LX, Kang G, Kumar P, Lu W, Li Y, Zhou Y, et al. (February 2014). "Humanized-BLT mouse model of Kaposi's sarcoma-associated herpesvirus infection". Proceedings of the National Academy of Sciences of the United States of America. 111 (8): 3146–51. Bibcode:2014PNAS..111.3146W. doi: 10.1073/pnas.1318175111 . PMC   3939909 . PMID   24516154.
  21. Wege AK, Florian C, Ernst W, Zimara N, Schleicher U, Hanses F, et al. (2012-07-24). "Leishmania major infection in humanized mice induces systemic infection and provokes a nonprotective human immune response". PLOS Neglected Tropical Diseases. 6 (7): e1741. doi: 10.1371/journal.pntd.0001741 . PMC   3404120 . PMID   22848771. S2CID   7657105.
  22. Amaladoss A, Chen Q, Liu M, Dummler SK, Dao M, Suresh S, et al. (2015-06-22). "De Novo Generated Human Red Blood Cells in Humanized Mice Support Plasmodium falciparum Infection". PLOS ONE. 10 (6): e0129825. Bibcode:2015PLoSO..1029825A. doi: 10.1371/journal.pone.0129825 . PMC   4476714 . PMID   26098918. S2CID   543860.
  23. Calderon VE, Valbuena G, Goez Y, Judy BM, Huante MB, Sutjita P, et al. (2013-05-17). "A humanized mouse model of tuberculosis". PLOS ONE. 8 (5): e63331. Bibcode:2013PLoSO...863331C. doi: 10.1371/journal.pone.0063331 . PMC   3656943 . PMID   23691024. S2CID   17215038.
  24. Frias-Staheli N, Dorner M, Marukian S, Billerbeck E, Labitt RN, Rice CM, Ploss A (February 2014). "Utility of humanized BLT mice for analysis of dengue virus infection and antiviral drug testing". Journal of Virology. 88 (4): 2205–18. doi:10.1128/JVI.03085-13. PMC   3911540 . PMID   24335303.
  25. Moffat JF, Stein MD, Kaneshima H, Arvin AM (September 1995). "Tropism of varicella-zoster virus for human CD4+ and CD8+ T lymphocytes and epidermal cells in SCID-hu mice". Journal of Virology. 69 (9): 5236–42. doi:10.1128/jvi.69.9.5236-5242.1995. PMC   189355 . PMID   7636965.
  26. Zheng J, Wu WL, Liu Y, Xiang Z, Liu M, Chan KH, et al. (2015-08-18). "The Therapeutic Effect of Pamidronate on Lethal Avian Influenza A H7N9 Virus Infected Humanized Mice". PLOS ONE. 10 (8): e0135999. Bibcode:2015PLoSO..1035999Z. doi: 10.1371/journal.pone.0135999 . PMC   4540487 . PMID   26285203. S2CID   10461525.
  27. 1 2 Tian H, Lyu Y, Yang YG, Hu Z (2020). "Humanized Rodent Models for Cancer Research". Frontiers in Oncology. 10: 1696. doi: 10.3389/fonc.2020.01696 . ISSN   2234-943X. PMC   7518015 . PMID   33042811. S2CID   221589508.
  28. Hausser HJ, Brenner RE (July 2005). "Phenotypic instability of Saos-2 cells in long-term culture". Biochemical and Biophysical Research Communications. 333 (1): 216–22. doi:10.1016/j.bbrc.2005.05.097. PMID   15939397.
  29. Wege AK, Schmidt M, Ueberham E, Ponnath M, Ortmann O, Brockhoff G, Lehmann J (2014-05-08). "Co-transplantation of human hematopoietic stem cells and human breast cancer cells in NSG mice: a novel approach to generate tumor cell specific human antibodies". mAbs. 6 (4): 968–77. doi:10.4161/mabs.29111. PMC   4171030 . PMID   24870377. S2CID   34234807.
  30. Her Z, Yong KS, Paramasivam K, Tan WW, Chan XY, Tan SY, et al. (October 2017). "An improved pre-clinical patient-derived liquid xenograft mouse model for acute myeloid leukemia". Journal of Hematology & Oncology. 10 (1): 162. doi: 10.1186/s13045-017-0532-x . PMC   5639594 . PMID   28985760.
  31. Her Z, Yong KS, Paramasivam K, Tan WW, Chan XY, Tan SY, et al. (October 2017). "An improved pre-clinical patient-derived liquid xenograft mouse model for acute myeloid leukemia". Journal of Hematology & Oncology. 10 (1): 162. doi: 10.1186/s13045-017-0532-x . PMC   5639594 . PMID   28985760. S2CID   25506701.
  32. Chen Q, Wang J, Liu WN, Zhao Y (July 2019). "Cancer Immunotherapies and Humanized Mouse Drug Testing Platforms". Translational Oncology. 12 (7): 987–995. doi:10.1016/j.tranon.2019.04.020. PMC   6529825 . PMID   31121491.
  33. Zayoud M, El Malki K, Frauenknecht K, Trinschek B, Kloos L, Karram K, et al. (September 2013). "Subclinical CNS inflammation as response to a myelin antigen in humanized mice". Journal of Neuroimmune Pharmacology. 8 (4): 1037–47. doi:10.1007/s11481-013-9466-4. PMID   23640521. S2CID   503830.
  34. Gunawan M, Her Z, Liu M, Tan SY, Chan XY, Tan WW, et al. (November 2017). "A Novel Human Systemic Lupus Erythematosus Model in Humanised Mice". Scientific Reports. 7 (1): 16642. Bibcode:2017NatSR...716642G. doi:10.1038/s41598-017-16999-7. PMC   5709358 . PMID   29192160. S2CID   5604139.
  35. King M, Pearson T, Shultz LD, Leif J, Bottino R, Trucco M, et al. (March 2008). "A new Hu-PBL model for the study of human islet alloreactivity based on NOD-scid mice bearing a targeted mutation in the IL-2 receptor gamma chain gene". Clinical Immunology. 126 (3): 303–314. doi:10.1016/j.clim.2007.11.001. PMID   18096436.

Further reading