1
|
Wang TT, Hirons A, Doerflinger M, Morris KV, Ledger S, Purcell DFJ, Kelleher AD, Ahlenstiel CL. Current State of Therapeutics for HTLV-1. Viruses 2024; 16:1616. [PMID: 39459949 PMCID: PMC11512412 DOI: 10.3390/v16101616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 10/07/2024] [Accepted: 10/11/2024] [Indexed: 10/28/2024] Open
Abstract
Human T cell leukaemia virus type-1 (HTLV-1) is an oncogenic retrovirus that causes lifelong infection in ~5-10 million individuals globally. It is endemic to certain First Nations populations of Northern and Central Australia, Japan, South and Central America, Africa, and the Caribbean region. HTLV-1 preferentially infects CD4+ T cells and remains in a state of reduced transcription, often being asymptomatic in the beginning of infection, with symptoms developing later in life. HTLV-1 infection is implicated in the development of adult T cell leukaemia/lymphoma (ATL) and HTLV-1-associated myelopathies (HAM), amongst other immune-related disorders. With no preventive or curative interventions, infected individuals have limited treatment options, most of which manage symptoms. The clinical burden and lack of treatment options directs the need for alternative treatment strategies for HTLV-1 infection. Recent advances have been made in the development of RNA-based antiviral therapeutics for Human Immunodeficiency Virus Type-1 (HIV-1), an analogous retrovirus that shares modes of transmission with HTLV-1. This review highlights past and ongoing efforts in the development of HTLV-1 therapeutics and vaccines, with a focus on the potential for gene therapy as a new treatment modality in light of its successes in HIV-1, as well as animal models that may help the advancement of novel antiviral and anticancer interventions.
Collapse
Affiliation(s)
- Tiana T. Wang
- Kirby Institute, University of New South Wales, Sydney, NSW 2052, Australia; (T.T.W.); (S.L.); (A.D.K.)
| | - Ashley Hirons
- The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC 3052, Australia; (A.H.); (D.F.J.P.)
| | - Marcel Doerflinger
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia;
- Department of Medical Biology, University of Melbourne, Melbourne, VIC 3050, Australia
| | - Kevin V. Morris
- Centre for Genomics and Personalised Health, School of Biomedical Sciences, Queensland University of Technology, Kelvin Grove, QLD 4059, Australia;
| | - Scott Ledger
- Kirby Institute, University of New South Wales, Sydney, NSW 2052, Australia; (T.T.W.); (S.L.); (A.D.K.)
| | - Damian F. J. Purcell
- The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC 3052, Australia; (A.H.); (D.F.J.P.)
| | - Anthony D. Kelleher
- Kirby Institute, University of New South Wales, Sydney, NSW 2052, Australia; (T.T.W.); (S.L.); (A.D.K.)
- UNSW RNA Institute, University of New South Wales, Sydney, NSW 2052, Australia
| | - Chantelle L. Ahlenstiel
- Kirby Institute, University of New South Wales, Sydney, NSW 2052, Australia; (T.T.W.); (S.L.); (A.D.K.)
- UNSW RNA Institute, University of New South Wales, Sydney, NSW 2052, Australia
| |
Collapse
|
2
|
Standley A, Xie J, Lau AW, Grote L, Gifford AJ. Working with Miraculous Mice: Mus musculus as a Model Organism. Curr Protoc 2024; 4:e70021. [PMID: 39435766 DOI: 10.1002/cpz1.70021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2024]
Abstract
The laboratory mouse has been described as a "miracle" model organism, providing a window by which we may gain an understanding of ourselves. Since the first recorded mouse experiment in 1664, the mouse has become the most used animal model in biomedical research. Mice are ideally suited as a model organism because of their small size, short gestation period, large litter size, and genetic similarity to humans. This article provides a broad overview of the laboratory mouse as a model organism and is intended for undergraduates and those new to working with mice. We delve into the history of the laboratory mouse and outline important terminology to accurately describe research mice. The types of laboratory mice available to researchers are reviewed, including outbred stocks, inbred strains, immunocompromised mice, and genetically engineered mice. The critical role mice have played in advancing knowledge in the areas of oncology, immunology, and pharmacology is highlighted by examining the significant contribution of mice to Nobel Prize winning research. International mouse mutagenesis programs and accurate phenotyping of mouse models are outlined. We also explain important considerations for working with mice, including animal ethics; the welfare principles of replacement, refinement, and reduction; and the choice of mouse model in experimental design. Finally, we present practical advice for maintaining a mouse colony, which involves adequate training of staff, the logistics of mouse housing, monitoring colony health, and breeding strategies. Useful resources for working with mice are also listed. The aim of this overview is to equip the reader with a broad appreciation of the enormous potential and some of the complexities of working with the laboratory mouse in a quest to improve human health. © 2024 The Author(s). Current Protocols published by Wiley Periodicals LLC.
Collapse
Affiliation(s)
- Anick Standley
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia
| | - Jinhan Xie
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia
| | - Angelica Wy Lau
- Garvan Institute of Medical Research, St Vincent's Clinical School, Darlinghurst, NSW, Australia
| | - Lauren Grote
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia
| | - Andrew J Gifford
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia
- Anatomical Pathology, NSW Heath Pathology, Prince of Wales Hospital, Randwick, NSW, Australia
- School of Clinical Medicine, UNSW Medicine & Health, UNSW Sydney, Sydney, NSW, Australia
| |
Collapse
|
3
|
Pascarella G, Conner KN, Goff NJ, Carninci P, Olive AJ, Meek K. Compared to other NHEJ factors, DNA-PK protein and RNA levels are markedly increased in all higher primates, but not in prosimians or other mammals. DNA Repair (Amst) 2024; 142:103737. [PMID: 39128395 PMCID: PMC11515020 DOI: 10.1016/j.dnarep.2024.103737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 07/23/2024] [Accepted: 07/24/2024] [Indexed: 08/13/2024]
Abstract
The DNA dependent protein kinase (DNA-PK) initiates non-homologous recombination (NHEJ), the predominate DNA double-strand break (DSBR) pathway in higher vertebrates. It has been known for decades that the enzymatic activity of DNA-PK [that requires its three component polypeptides, Ku70, Ku80 (that comprise the DNA-end binding Ku heterodimer), and the catalytic subunit (DNA-PKcs)] is present in humans at 10-50 times the level observed in other mammals. Here, we show that the high level of DNA-PKcs protein expression appears evolutionarily in mammals between prosimians and higher primates. Moreover, the RNAs encoding the three component polypeptides of DNA-PK are present at similarly high levels in hominids, new-, and old-world monkeys, but expression of these RNAs in prosimians is ∼5-50 fold less, analogous to the levels observed in other non-primate species. This is reminiscent of the appearance of Alu repeats in primate genomes -- abundant in higher primates, but present at much lower density in prosimians. Alu repeats are well-known for their capacity to promote non-allelic homologous recombination (NAHR) a process known to be inhibited by DNA-PK. Nanopore sequence analyses of cultured cells proficient or deficient in DNA-PK revealed an increase of inter-chromosomal translocations caused by NAHR. Although the high levels of DNA-PK in primates may have many functions, we posit that high levels of DNA-PK may function to restrain deleterious NAHR events between Alu elements.
Collapse
Affiliation(s)
| | - Kayla N Conner
- Department of Microbiology, Genetics, and Immunology, Michigan State University, East Lansing, MI 48824, USA
| | - Noah J Goff
- Department of Microbiology, Genetics, and Immunology, Michigan State University, East Lansing, MI 48824, USA; Department of Pathobiology & Diagnostic Investigation, Michigan State University, East Lansing, MI 48824, USA
| | - Piero Carninci
- RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Japan; Human Technopole, Milan, Italy
| | - Andrew J Olive
- Department of Microbiology, Genetics, and Immunology, Michigan State University, East Lansing, MI 48824, USA
| | - Katheryn Meek
- Department of Microbiology, Genetics, and Immunology, Michigan State University, East Lansing, MI 48824, USA; Department of Pathobiology & Diagnostic Investigation, Michigan State University, East Lansing, MI 48824, USA.
| |
Collapse
|
4
|
Guzman RM, Voth DE. Embracing multiple infection models to tackle Q fever: A review of in vitro, in vivo, and lung ex vivo models. Cell Immunol 2024; 405-406:104880. [PMID: 39357100 DOI: 10.1016/j.cellimm.2024.104880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 09/06/2024] [Accepted: 09/23/2024] [Indexed: 10/04/2024]
Abstract
Multiple animal and cell culture models are employed to study pathogenesis of Coxiella burnetii, the causative agent of acute and chronic human Q fever. C. burnetii is a lung pathogen that is aerosolized in contaminated products and inhaled by humans to cause acute disease that can disseminate to other organs and establish chronic infection. Cellular models of Q fever include a variety of tissue-derived cell lines from mice and humans such as lung alveolar ex vivo cells. These models have the advantage of being cost-effective and reproducible. Similarly, animal models including mice and guinea pigs are cost-effective, although only immunocompromised SCID mice display a severe disease phenotype in response to Nine Mile I and Nine Mile II isolates of C. burnetii while immunocompetent guinea pigs display human-like symptoms and robust immune responses. Non-human primates such as macaques and marmosets are the closest model of human disease but are costly and largely used for adaptive immune response studies. All animal models are used for vaccine development but many differences exist in the pathogen's ability to establish lung infection when considering infection routes, bacterial isolates, and host genetic background. Similarly, while cellular models are useful for characterization of host-pathogen mechanisms, future developments should include use of a lung infection platform to draw appropriate conclusions. Here, we summarize the current state of the C. burnetii lung pathogenesis field by discussing the contribution of different animal and cell culture models and include suggestions for continuing to move the field forward.
Collapse
Affiliation(s)
- R M Guzman
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States
| | - Daniel E Voth
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States.
| |
Collapse
|
5
|
Yadav KK, Kenney SP. Hepatitis E virus immunosuppressed animal models. BMC Infect Dis 2024; 24:965. [PMID: 39266958 PMCID: PMC11395946 DOI: 10.1186/s12879-024-09870-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 09/03/2024] [Indexed: 09/14/2024] Open
Abstract
Hepatitis E virus (HEV) is an important emerging pathogen producing significant morbidity in immunosuppressed patients. HEV has been detrimental to solid organ transplant (SOT) patients, cancer patients, and HIV-positive patients, where chronic HEV infections occur. Blood-borne transfusions and multiple cases of chronic HEV infection in transplant patients have been reported in the past few decades, necessitating research on HEV pathogenesis using immunosuppressed animal models. Numerous animal species with unique naturally occurring HEV strains have been found, several of which have the potential to spread to humans and to serve as pathogenesis models. Host immunosuppression leads to viral persistence and chronic HEV infection allows for genetic adaptation to the human host creating new strains with worse disease outcomes. Procedures necessary for SOT often entail blood transfusions placing immunosuppressive patients into a "high risk group" for HEV infection. This scenario requires an appropriate immunosuppressive animal model to understand disease patterns in these patients. Hence, this article reviews the recent advances in the immunosuppressed animal models for chronic HEV infection with emphasis on pathogenesis, immune correlates, and the liver pathology associated with the chronic HEV infections.
Collapse
Affiliation(s)
- Kush Kumar Yadav
- Center for Food Animal Health, Department of Animal Sciences, The Ohio State University, 1680 Madison Ave, Wooster, OH, 44691, USA
- Department of Veterinary Preventive Medicine, The Ohio State University, Columbus, 43210, USA
| | - Scott P Kenney
- Center for Food Animal Health, Department of Animal Sciences, The Ohio State University, 1680 Madison Ave, Wooster, OH, 44691, USA.
- Department of Veterinary Preventive Medicine, The Ohio State University, Columbus, 43210, USA.
| |
Collapse
|
6
|
Vandecandelaere G, Ramapriyan R, Gaffey M, Richardson LG, Steuart SJ, Tazhibi M, Kalaw A, Grewal EP, Sun J, Curry WT, Choi BD. Pre-Clinical Models for CAR T-Cell Therapy for Glioma. Cells 2024; 13:1480. [PMID: 39273050 PMCID: PMC11394304 DOI: 10.3390/cells13171480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 08/28/2024] [Accepted: 09/01/2024] [Indexed: 09/15/2024] Open
Abstract
Immunotherapy represents a transformative shift in cancer treatment. Among myriad immune-based approaches, chimeric antigen receptor (CAR) T-cell therapy has shown promising results in treating hematological malignancies. Despite aggressive treatment options, the prognosis for patients with malignant brain tumors remains poor. Research leveraging CAR T-cell therapy for brain tumors has surged in recent years. Pre-clinical models are crucial in evaluating the safety and efficacy of these therapies before they advance to clinical trials. However, current models recapitulate the human tumor environment to varying degrees. Novel in vitro and in vivo techniques offer the opportunity to validate CAR T-cell therapies but also have limitations. By evaluating the strengths and weaknesses of various pre-clinical glioma models, this review aims to provide a roadmap for the development and pre-clinical testing of CAR T-cell therapies for brain tumors.
Collapse
Affiliation(s)
- Gust Vandecandelaere
- Brain Tumor Immunotherapy Lab, Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; (G.V.)
- Faculty of Medicine, KU Leuven, 3000 Leuven, Belgium
| | - Rishab Ramapriyan
- Brain Tumor Immunotherapy Lab, Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; (G.V.)
| | - Matthew Gaffey
- Brain Tumor Immunotherapy Lab, Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; (G.V.)
| | - Leland Geoffrey Richardson
- Brain Tumor Immunotherapy Lab, Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; (G.V.)
| | - Samuel Jeffrey Steuart
- Brain Tumor Immunotherapy Lab, Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; (G.V.)
| | - Masih Tazhibi
- Brain Tumor Immunotherapy Lab, Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; (G.V.)
| | - Adrian Kalaw
- Brain Tumor Immunotherapy Lab, Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; (G.V.)
| | - Eric P. Grewal
- Brain Tumor Immunotherapy Lab, Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; (G.V.)
| | - Jing Sun
- Brain Tumor Immunotherapy Lab, Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; (G.V.)
| | - William T. Curry
- Brain Tumor Immunotherapy Lab, Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; (G.V.)
| | - Bryan D. Choi
- Brain Tumor Immunotherapy Lab, Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; (G.V.)
| |
Collapse
|
7
|
Mizuno M, Kiyotake S, Matsubayashi M, Kaneko T, Hatai H, Fujimoto Y, Ijiri M, Kawaguchi H, Matsui T, Matsuo T. Potential Development Ability of Residual Zoites, a Second-Generation Meront, Inducing Long-Term Infection by the Mouse Eimerian Parasite, Eimeria krijgsmanni. Acta Parasitol 2024:10.1007/s11686-024-00910-2. [PMID: 39207650 DOI: 10.1007/s11686-024-00910-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 08/13/2024] [Indexed: 09/04/2024]
Abstract
PURPOSE Coccidiosis caused by eimerian parasites results in lethal watery or bloody diarrhea in hosts, and markedly impairs the growth of and feed utilization by host animals. We previously investigated detailed the life cycle of Eimeria krijgsmanni as a mouse eimerian parasite. Only second-generation meronts, as an asexual stage, were morphologically detected in the epithelium of the host cecum for at least 8 weeks after infection, even though oocyst shedding finished approximately 3 weeks after infection. The presence of zoites was of interest because infection by eimerian parasites is considered to be self-limited after their patent period. METHODS To clarify the significance of residual second-generation meronts in E. krijgsmanni infection, we performed infection experiments using immunocompetent mice under artificial immunosuppression and congenital immunodeficient mice. RESULTS The number of oocysts discharged and the duration of oocyst discharge both increased in immunosuppressed mice. In immunodeficient mice, numerous oocysts were shed over a markedly longer period, and oocyst discharge did not finish until 56 days after inoculation. CONCLUSIONS The present results suggest that the second-generation meronts survived in the epithelial cells of the cecum after the patent period, thereby contributing to extended infection as an asexual stage. The results obtained on E. krijgsmanni indicate that infections by Eimeria spp. are not self-limited and potentially continue for a long period of time.
Collapse
Affiliation(s)
- Masanobu Mizuno
- Laboratory of Parasitology, Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima, 890-0065, Japan
| | - Satoru Kiyotake
- Laboratory of Parasitology, Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima, 890-0065, Japan
| | - Makoto Matsubayashi
- Laboratory of Veterinary Immunology, Graduate School of Veterinary Science, Osaka Metropolitan University, Izumisano, 598-5831, Osaka, Japan
| | - Takane Kaneko
- Department of Life Sciences, Faculty of Life Sciences, Kyushu Sangyo University, Fukuoka, 813-8503, Japan
| | - Hitoshi Hatai
- Farm Animal Clinical Skills and Diseases Control Center, Iwate University, Morioka, 020-8550, Iwate, Japan
| | - Yoshikazu Fujimoto
- Transboundary Animal Diseases Research Center, Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima, 890-0065, Japan
| | - Moe Ijiri
- Transboundary Animal Diseases Research Center, Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima, 890-0065, Japan
| | - Hiroaki Kawaguchi
- Laboratory of Veterinary Pathology, School of Veterinary Medicine, Kitasato University, Towada, 034-8628, Aomori, Japan
| | | | - Tomohide Matsuo
- Laboratory of Parasitology, Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima, 890-0065, Japan.
| |
Collapse
|
8
|
Kwak H, Lee E, Karki R. DNA sensors in metabolic and cardiovascular diseases: Molecular mechanisms and therapeutic prospects. Immunol Rev 2024. [PMID: 39158380 DOI: 10.1111/imr.13382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/20/2024]
Abstract
DNA sensors generally initiate innate immune responses through the production of type I interferons. While extensively studied for host defense against invading pathogens, emerging evidence highlights the involvement of DNA sensors in metabolic and cardiovascular diseases. Elevated levels of modified, damaged, or ectopically localized self-DNA and non-self-DNA have been observed in patients and animal models with obesity, diabetes, fatty liver disease, and cardiovascular disease. The accumulation of cytosolic DNA aberrantly activates DNA signaling pathways, driving the pathological progression of these disorders. This review highlights the roles of specific DNA sensors, such as cyclic AMP-GMP synthase and stimulator of interferon genes (cGAS-STING), absent in melanoma 2 (AIM2), toll-like receptor 9 (TLR9), interferon gamma-inducible protein 16 (IFI16), DNA-dependent protein kinase (DNA-PK), and DEAD-box helicase 41 (DDX41) in various metabolic disorders. We explore how DNA signaling pathways in both immune and non-immune cells contribute to the development of these diseases. Furthermore, we discuss the intricate interplay between metabolic stress and immune responses, offering insights into potential therapeutic targets for managing metabolic and cardiovascular disorders. Understanding the mechanisms of DNA sensor signaling in these contexts provides a foundation for developing novel interventions aimed at mitigating the impact of these pervasive health issues.
Collapse
Affiliation(s)
- Hyosang Kwak
- Department of Biological Sciences, College of Natural Science, Seoul National University, Seoul, South Korea
| | - Ein Lee
- Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, South Korea
| | - Rajendra Karki
- Department of Biological Sciences, College of Natural Science, Seoul National University, Seoul, South Korea
- Nexus Institute of Research and Innovation (NIRI), Kathmandu, Nepal
| |
Collapse
|
9
|
Xiao Q, Zhai L, Zhang X, Liu Y, Li J, Xie X, Xu G, He S, Fu H, Tang Y, Zhang F, Liu Y. How can we establish animal models of HIV-associated lymphoma? Animal Model Exp Med 2024; 7:484-496. [PMID: 38567763 PMCID: PMC11369037 DOI: 10.1002/ame2.12409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 02/27/2024] [Indexed: 09/04/2024] Open
Abstract
Human immunodeficiency virus (HIV) infection is strongly associated with a heightened incidence of lymphomas. To mirror the natural course of human HIV infection, animal models have been developed. These models serve as valuable tools to investigate disease pathobiology, assess antiretroviral and immunomodulatory drugs, explore viral reservoirs, and develop eradication strategies. However, there are currently no validated in vivo models of HIV-associated lymphoma (HAL), hampering progress in this crucial domain, and scant attention has been given to developing animal models dedicated to studying HAL, despite their pivotal role in advancing knowledge. This review provides a comprehensive overview of the existing animal models of HAL, which may enhance our understanding of the underlying pathogenesis and approaches for malignancies linked to HIV infection.
Collapse
Affiliation(s)
- Qing Xiao
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Department of Hematology‐OncologyChongqing University Cancer HospitalChongqingChina
| | - Liuyue Zhai
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Department of Hematology‐OncologyChongqing University Cancer HospitalChongqingChina
| | - Xiaomei Zhang
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Department of Hematology‐OncologyChongqing University Cancer HospitalChongqingChina
| | - Yi Liu
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Department of Hematology‐OncologyChongqing University Cancer HospitalChongqingChina
| | - Jun Li
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Department of Hematology‐OncologyChongqing University Cancer HospitalChongqingChina
| | - Xiaoqing Xie
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Department of Hematology‐OncologyChongqing University Cancer HospitalChongqingChina
| | - Guofa Xu
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Department of Hematology‐OncologyChongqing University Cancer HospitalChongqingChina
| | - Sanxiu He
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Department of Hematology‐OncologyChongqing University Cancer HospitalChongqingChina
| | - Huihui Fu
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Department of Hematology‐OncologyChongqing University Cancer HospitalChongqingChina
| | - Yifeng Tang
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Department of Hematology‐OncologyChongqing University Cancer HospitalChongqingChina
| | - Fujie Zhang
- Beijing Ditan HospitalCapital Medical UniversityBeijingChina
| | - Yao Liu
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Department of Hematology‐OncologyChongqing University Cancer HospitalChongqingChina
| |
Collapse
|
10
|
Bin Y, Ren J, Zhang H, Zhang T, Liu P, Xin Z, Yang H, Feng Z, Chen Z, Zhang H. Against all odds: The road to success in the development of human immune reconstitution mice. Animal Model Exp Med 2024; 7:460-470. [PMID: 38591343 PMCID: PMC11369039 DOI: 10.1002/ame2.12407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 03/17/2024] [Indexed: 04/10/2024] Open
Abstract
The mouse genome has a high degree of homology with the human genome, and its physiological, biochemical, and developmental regulation mechanisms are similar to those of humans; therefore, mice are widely used as experimental animals. However, it is undeniable that interspecies differences between humans and mice can lead to experimental errors. The differences in the immune system have become an important factor limiting current immunological research. The application of immunodeficient mice provides a possible solution to these problems. By transplanting human immune cells or tissues, such as peripheral blood mononuclear cells or hematopoietic stem cells, into immunodeficient mice, a human immune system can be reconstituted in the mouse body, and the engrafted immune cells can elicit human-specific immune responses. Researchers have been actively exploring the development and differentiation conditions of host recipient animals and grafts in order to achieve better immune reconstitution. Through genetic engineering methods, immunodeficient mice can be further modified to provide a favorable developmental and differentiation microenvironment for the grafts. From initially only being able to reconstruct single T lymphocyte lineages, it is now possible to reconstruct lymphoid and myeloid cells, providing important research tools for immunology-related studies. In this review, we compare the differences in immune systems of humans and mice, describe the development history of human immune reconstitution from the perspectives of immunodeficient mice and grafts, and discuss the latest advances in enhancing the efficiency of human immune cell reconstitution, aiming to provide important references for immunological related researches.
Collapse
Affiliation(s)
- Yixiao Bin
- School of Basic Medical SciencesShaanxi University of Chinese MedicineXianyangChina
- Department of Cell Biology, National Translational Science Center for Molecular MedicineFourth Military Medical UniversityXi'anChina
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesFourth Military Medical UniversityXi'anChina
| | - Jing Ren
- School of Basic Medical SciencesShaanxi University of Chinese MedicineXianyangChina
- Department of Cell Biology, National Translational Science Center for Molecular MedicineFourth Military Medical UniversityXi'anChina
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesFourth Military Medical UniversityXi'anChina
| | - Haowei Zhang
- Department of Occupational & Environmental Health and the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public HealthFourth Military Medical UniversityXi'anChina
| | - Tianjiao Zhang
- Department of Cell Biology, National Translational Science Center for Molecular MedicineFourth Military Medical UniversityXi'anChina
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesFourth Military Medical UniversityXi'anChina
| | - Peijuan Liu
- Department of Cell Biology, National Translational Science Center for Molecular MedicineFourth Military Medical UniversityXi'anChina
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesFourth Military Medical UniversityXi'anChina
| | - Zhiqian Xin
- Department of Cell Biology, National Translational Science Center for Molecular MedicineFourth Military Medical UniversityXi'anChina
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesFourth Military Medical UniversityXi'anChina
| | - Haijiao Yang
- Department of Cell Biology, National Translational Science Center for Molecular MedicineFourth Military Medical UniversityXi'anChina
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesFourth Military Medical UniversityXi'anChina
| | - Zhuan Feng
- Department of Cell Biology, National Translational Science Center for Molecular MedicineFourth Military Medical UniversityXi'anChina
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesFourth Military Medical UniversityXi'anChina
| | - Zhinan Chen
- Department of Cell Biology, National Translational Science Center for Molecular MedicineFourth Military Medical UniversityXi'anChina
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesFourth Military Medical UniversityXi'anChina
| | - Hai Zhang
- Department of Cell Biology, National Translational Science Center for Molecular MedicineFourth Military Medical UniversityXi'anChina
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesFourth Military Medical UniversityXi'anChina
| |
Collapse
|
11
|
Chou TC, Maggirwar NS, Marsden MD. HIV Persistence, Latency, and Cure Approaches: Where Are We Now? Viruses 2024; 16:1163. [PMID: 39066325 PMCID: PMC11281696 DOI: 10.3390/v16071163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 07/13/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
The latent reservoir remains a major roadblock to curing human immunodeficiency virus (HIV) infection. Currently available antiretroviral therapy (ART) can suppress active HIV replication, reduce viral loads to undetectable levels, and halt disease progression. However, antiretroviral drugs are unable to target cells that are latently infected with HIV, which can seed viral rebound if ART is stopped. Consequently, a major focus of the field is to study the latent viral reservoir and develop safe and effective methods to eliminate it. Here, we provide an overview of the major mechanisms governing the establishment and maintenance of HIV latency, the key challenges posed by latent reservoirs, small animal models utilized to study HIV latency, and contemporary cure approaches. We also discuss ongoing efforts to apply these approaches in combination, with the goal of achieving a safe, effective, and scalable cure for HIV that can be extended to the tens of millions of people with HIV worldwide.
Collapse
Affiliation(s)
- Tessa C. Chou
- Department of Microbiology and Molecular Genetics, School of Medicine, University of California, Irvine, CA 92617, USA; (T.C.C.); (N.S.M.)
| | - Nishad S. Maggirwar
- Department of Microbiology and Molecular Genetics, School of Medicine, University of California, Irvine, CA 92617, USA; (T.C.C.); (N.S.M.)
| | - Matthew D. Marsden
- Department of Microbiology and Molecular Genetics, School of Medicine, University of California, Irvine, CA 92617, USA; (T.C.C.); (N.S.M.)
- Department of Medicine, Division of Infectious Disease, School of Medicine, University of California, Irvine, CA 92617, USA
| |
Collapse
|
12
|
McDonald K, Rodriguez A, Muthukrishnan G. Humanized Mouse Models of Bacterial Infections. Antibiotics (Basel) 2024; 13:640. [PMID: 39061322 PMCID: PMC11273811 DOI: 10.3390/antibiotics13070640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 07/02/2024] [Accepted: 07/06/2024] [Indexed: 07/28/2024] Open
Abstract
Bacterial infections continue to represent a significant healthcare burden worldwide, causing considerable mortality and morbidity every year. The emergence of multidrug-resistant bacterial strains continues to rise, posing serious risks to controlling global disease outbreaks. To develop novel and more effective treatment and vaccination programs, there is a need for clinically relevant small animal models. Since multiple bacterial species have human-specific tropism for numerous virulence factors and toxins, conventional mouse models do not fully represent human disease. Several human disease characteristic phenotypes, such as lung granulomas in the case of Mycobacterium tuberculosis infections, are absent in standard mouse models. Alternatively, certain pathogens, such as Salmonella enterica serovar typhi and Staphylococcus aureus, can be well tolerated in mice and cleared quickly. To address this, multiple groups have developed humanized mouse models and observed enhanced susceptibility to infection and a more faithful recapitulation of human disease. In the last two decades, multiple humanized mouse models have been developed to attempt to recapitulate the human immune system in a small animal model. In this review, we first discuss the history of immunodeficient mice that has enabled the engraftment of human tissue and the engraftment methods currently used in the field. We then highlight how humanized mouse models successfully uncovered critical human immune responses to various bacterial infections, including Salmonella enterica serovar Typhi, Mycobacterium tuberculosis, and Staphylococcus aureus.
Collapse
Affiliation(s)
- Katya McDonald
- Center for Musculoskeletal Research, Department of Orthopaedics, University of Rochester Medical Center, 601 Elmwood Avenue, Box 665, Rochester, NY 14642, USA
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Adryiana Rodriguez
- Center for Musculoskeletal Research, Department of Orthopaedics, University of Rochester Medical Center, 601 Elmwood Avenue, Box 665, Rochester, NY 14642, USA
| | - Gowrishankar Muthukrishnan
- Center for Musculoskeletal Research, Department of Orthopaedics, University of Rochester Medical Center, 601 Elmwood Avenue, Box 665, Rochester, NY 14642, USA
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA
| |
Collapse
|
13
|
Cocco E, de Stanchina E. Patient-Derived-Xenografts in Mice: A Preclinical Platform for Cancer Research. Cold Spring Harb Perspect Med 2024; 14:a041381. [PMID: 37696659 PMCID: PMC11216185 DOI: 10.1101/cshperspect.a041381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/13/2023]
Abstract
The use of patient-derived xenografts (PDXs) has dramatically improved drug development programs. PDXs (1) reproduce the pathological features and the genomic profile of the parental tumors more precisely than other preclinical models, and (2) more faithfully predict therapy response. However, PDXs have limitations. These include the inability to completely capture tumor heterogeneity and the role of the immune system, the low engraftment efficiency of certain tumor types, and the consequences of the human-host interactions. Recently, the use of novel mouse strains and specialized engraftment techniques has enabled the generation of "humanized" PDXs, partially overcoming such limitations. Importantly, establishing, characterizing, and maintaining PDXs is costly and requires a significant regulatory, administrative, clinical, and laboratory infrastructure. In this review, we will retrace the historical milestones that led to the implementation of PDXs for cancer research, review the most recent innovations in the field, and discuss future avenues to tackle deficiencies that still exist.
Collapse
Affiliation(s)
- Emiliano Cocco
- University of Miami, Miller School of Medicine, Department of Biochemistry and Molecular Biology, Sylvester Comprehensive Cancer Center, Miami, Florida 33136, USA
| | - Elisa de Stanchina
- Antitumor Assessment Core Facility, Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| |
Collapse
|
14
|
Raghunandanan S, Zhang K, Zhang Y, Sze CW, Priya R, Luo Y, Lynch MJ, Crane BR, Li C, Yang XF. MCP5, a methyl-accepting chemotaxis protein regulated by both the Hk1-Rrp1 and Rrp2-RpoN-RpoS pathways, is required for the immune evasion of Borrelia burgdorferi. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.10.598185. [PMID: 38915556 PMCID: PMC11195095 DOI: 10.1101/2024.06.10.598185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Borrelia (or Borreliella) burgdorferi, the causative agent of Lyme disease, is a motile and invasive zoonotic pathogen, adept at navigating between its arthropod vector and mammalian host. While motility and chemotaxis are well established as essential for its enzootic cycle, the function of methyl-accepting chemotaxis proteins (MCPs) in the infectious cycle of B. burgdorferi remains unclear. In this study, we demonstrate that MCP5, one of the most abundant MCPs in B. burgdorferi, is differentially expressed in response to environmental signals as well as at different stages of the pathogen's enzootic cycle. Specifically, the expression of mcp5 is regulated by the Hk1-Rrp1 and Rrp2-RpoN-RpoS pathways, which are critical for the spirochete's colonization of the tick vector and mammalian host, respectively. Infection experiments with an mcp5 mutant revealed that spirochetes lacking MCP5 could not establish infections in either C3H/HeN mice or Severe Combined Immunodeficiency (SCID) mice, which are defective in adaptive immunity, indicating the essential role of MCP5 in mammalian infection. However, the mcp5 mutant could establish infection and disseminate in NOD SCID Gamma (NSG) mice, which are deficient in both adaptive and most innate immune responses, suggesting a crucial role of MCP5 in evading host innate immunity. In the tick vector, the mcp5 mutants survived feeding but failed to transmit to mice, highlighting the importance of MCP5 in transmission. Our findings reveal that MCP5, regulated by the Rrp1 and Rrp2 pathways, is critical for the establishment of infection in mammalian hosts by evading host innate immunity and is important for the transmission of spirochetes from ticks to mammalian hosts, underscoring its potential as a target for intervention strategies.
Collapse
Affiliation(s)
- Sajith Raghunandanan
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Kai Zhang
- Department of Oral Craniofacial Molecular Biology, School of Dentistry, Virginia Commonwealth University, Richmond, Virginia, 23298
| | - Yan Zhang
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202
- Wenzhou Key Laboratory of Sanitary Microbiology, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China, 325035
| | - Ching Wooen Sze
- Department of Oral Craniofacial Molecular Biology, School of Dentistry, Virginia Commonwealth University, Richmond, Virginia, 23298
| | - Raj Priya
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Yongliang Luo
- Wenzhou Key Laboratory of Sanitary Microbiology, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China, 325035
| | - Michael J Lynch
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY 14853, USA
| | - Brian R Crane
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY 14853, USA
| | - Chunhao Li
- Department of Oral Craniofacial Molecular Biology, School of Dentistry, Virginia Commonwealth University, Richmond, Virginia, 23298
| | - X. Frank Yang
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202
| |
Collapse
|
15
|
Sahu SR, Dutta A, Peroumal D, Kumari P, Utakalaja BG, Patel SK, Acharya N. Immunogenicity and efficacy of CNA25 as a potential whole-cell vaccine against systemic candidiasis. EMBO Mol Med 2024; 16:1254-1283. [PMID: 38783167 PMCID: PMC11178797 DOI: 10.1038/s44321-024-00080-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 05/10/2024] [Accepted: 05/10/2024] [Indexed: 05/25/2024] Open
Abstract
Disseminated fungal infections account for ~1.5 million deaths per year worldwide, and mortality may increase further due to a rise in the number of immunocompromised individuals and drug-resistance fungal species. Since an approved antifungal vaccine is yet to be available, this study explored the immunogenicity and vaccine efficacy of a DNA polymerase mutant strain of Candida albicans. CNA25 is a pol32ΔΔ strain that exhibits growth defects and does not cause systemic candidiasis in mice. Immunized mice with live CNA25 were fully protected against C. albicans and C. parapsilosis but partially against C. tropicalis and C. glabrata infections. CNA25 induced steady expression of TLR2 and Dectin-1 receptors leading to a faster recognition and clearance by the immune system associated with the activation of protective immune responses mostly mediated by neutrophils, macrophages, NK cells, B cells, and CD4+ and CD8+ T cells. Molecular blockade of Dectin-1, IL-17, IFNγ, and TNFα abolished resistance to reinfection. Altogether, this study suggested that CNA25 collectively activates innate, adaptive, and trained immunity to be a promising live whole-cell vaccine against systemic candidiasis.
Collapse
Affiliation(s)
- Satya Ranjan Sahu
- Department of Infectious Disease Biology, Institute of Life Sciences, Bhubaneswar, Odisha, 751023, India
- Regional Center for Biotechnology, Faridabad, Haryana, 751021, India
| | - Abinash Dutta
- Department of Infectious Disease Biology, Institute of Life Sciences, Bhubaneswar, Odisha, 751023, India
| | - Doureradjou Peroumal
- Department of Infectious Disease Biology, Institute of Life Sciences, Bhubaneswar, Odisha, 751023, India
| | - Premlata Kumari
- Department of Infectious Disease Biology, Institute of Life Sciences, Bhubaneswar, Odisha, 751023, India
- Regional Center for Biotechnology, Faridabad, Haryana, 751021, India
| | - Bhabasha Gyanadeep Utakalaja
- Department of Infectious Disease Biology, Institute of Life Sciences, Bhubaneswar, Odisha, 751023, India
- Regional Center for Biotechnology, Faridabad, Haryana, 751021, India
| | - Shraddheya Kumar Patel
- Department of Infectious Disease Biology, Institute of Life Sciences, Bhubaneswar, Odisha, 751023, India
- Regional Center for Biotechnology, Faridabad, Haryana, 751021, India
| | - Narottam Acharya
- Department of Infectious Disease Biology, Institute of Life Sciences, Bhubaneswar, Odisha, 751023, India.
| |
Collapse
|
16
|
Hasler MF, Speck RF, Kadzioch NP. Humanized mice for studying HIV latency and potentially its eradication. Curr Opin HIV AIDS 2024; 19:157-167. [PMID: 38547338 DOI: 10.1097/coh.0000000000000855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
PURPOSE OF THE REVIEW The quest for an HIV cure faces a formidable challenge: the persistent presence of latent viral infections within the cells and tissues of infected individuals. This review provides a thorough examination of discussions surrounding HIV latency, the use of humanized mouse models, and strategies aimed at eliminating the latent HIV reservoir. It explores the hurdles and advancements in understanding HIV pathogenesis, mainly focusing on establishing latent reservoirs in CD4 + T cells and macrophages. Introducing the concepts of functional and sterile cures, the review underscores the indispensable role of humanized mouse models in HIV research, offering crucial insights into the efficacy of cART and the ongoing pursuit of an HIV cure. RECENT FINDINGS Here, we highlight studies investigating molecular mechanisms and pathogenesis related to HIV latency in humanized mice and discuss novel strategies for eradicating latent HIV. Emphasizing the importance of analytical cART interruption in humanized mouse studies to gauge its impact on the latent reservoir accurately, the review underlines the ongoing progress and challenges in harnessing humanized mouse models for HIV research. SUMMARY This review suggests that humanized mice models provide valuable insights into HIV latency and potential eradication strategies, contributing significantly to the quest for an HIV cure.
Collapse
Affiliation(s)
- Moa F Hasler
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital of Zurich, University of Zurich, Zurich, Switzerland
| | | | | |
Collapse
|
17
|
De Meyer A, Meuleman P. Preclinical animal models to evaluate therapeutic antiviral antibodies. Antiviral Res 2024; 225:105843. [PMID: 38548022 DOI: 10.1016/j.antiviral.2024.105843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 02/25/2024] [Indexed: 04/05/2024]
Abstract
Despite the availability of effective preventative vaccines and potent small-molecule antiviral drugs, effective non-toxic prophylactic and therapeutic measures are still lacking for many viruses. The use of monoclonal and polyclonal antibodies in an antiviral context could fill this gap and provide effective virus-specific medical interventions. In order to develop these therapeutic antibodies, preclinical animal models are of utmost importance. Due to the variability in viral pathogenesis, immunity and overall characteristics, the most representative animal model for human viral infection differs between virus species. Therefore, throughout the years researchers sought to find the ideal preclinical animal model for each virus. The most used animal models in preclinical research include rodents (mice, ferrets, …) and non-human primates (macaques, chimpanzee, ….). Currently, antibodies are tested for antiviral efficacy against a variety of viruses including different hepatitis viruses, human immunodeficiency virus (HIV), influenza viruses, respiratory syncytial virus (RSV), severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and rabies virus. This review provides an overview of the current knowledge about the preclinical animal models that are used for the evaluation of therapeutic antibodies for the abovementioned viruses.
Collapse
Affiliation(s)
- Amse De Meyer
- Laboratory of Liver Infectious Diseases, Department of Diagnostic Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Philip Meuleman
- Laboratory of Liver Infectious Diseases, Department of Diagnostic Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium.
| |
Collapse
|
18
|
Mina A, Pavletic S, Aplan PD. The evolution of preclinical models for myelodysplastic neoplasms. Leukemia 2024; 38:683-691. [PMID: 38396286 PMCID: PMC10997513 DOI: 10.1038/s41375-024-02181-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 02/06/2024] [Accepted: 02/08/2024] [Indexed: 02/25/2024]
Abstract
Myelodysplastic Neoplasms (MDS) are a group of clonal disorders characterized by ineffective hematopoiesis and morphologic dysplasia. Clinical manifestations of MDS vary widely and are dictated in large part by a range of genetic aberrations. The lack of robust in vitro models for MDS has limited the ability to conduct high throughput drug screens, which in turn has hampered the development of novel therapies for MDS. There are very few well-characterized MDS cell lines, and the available cell lines expand poorly in vitro. Conventional xenograft mouse models can provide an in vivo vessel to provide growth of cancer cells, but human MDS cells engraft poorly. Three-dimensional (3D) scaffold models that form human "ossicles" represent a promising new approach and can reproduce the intricate communication between hematopoietic stem and progenitor cells and their environment. Genetically engineered mice utilize specific mutations and may not represent the entire array of human MDS; however, genetically engineered mice provided in vivo proof of principle for novel agents such as luspatercept, demonstrating the clinical utility of this approach. This review offers an overview of available preclinical MDS models and potential approaches to accelerate accurate clinical translation.
Collapse
Affiliation(s)
- Alain Mina
- Myeloid Malignancies Program, Immune Deficiency Cellular Therapy Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| | - Steven Pavletic
- Myeloid Malignancies Program, Immune Deficiency Cellular Therapy Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Peter D Aplan
- Myeloid Malignancies Program, Immune Deficiency Cellular Therapy Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
19
|
Sun C, Serra C, Kalicharan BH, Harding J, Rao M. Challenges and Considerations of Preclinical Development for iPSC-Based Myogenic Cell Therapy. Cells 2024; 13:596. [PMID: 38607035 PMCID: PMC11011706 DOI: 10.3390/cells13070596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 03/21/2024] [Accepted: 03/22/2024] [Indexed: 04/13/2024] Open
Abstract
Cell therapies derived from induced pluripotent stem cells (iPSCs) offer a promising avenue in the field of regenerative medicine due to iPSCs' expandability, immune compatibility, and pluripotent potential. An increasing number of preclinical and clinical trials have been carried out, exploring the application of iPSC-based therapies for challenging diseases, such as muscular dystrophies. The unique syncytial nature of skeletal muscle allows stem/progenitor cells to integrate, forming new myonuclei and restoring the expression of genes affected by myopathies. This characteristic makes genome-editing techniques especially attractive in these therapies. With genetic modification and iPSC lineage specification methodologies, immune-compatible healthy iPSC-derived muscle cells can be manufactured to reverse the progression of muscle diseases or facilitate tissue regeneration. Despite this exciting advancement, much of the development of iPSC-based therapies for muscle diseases and tissue regeneration is limited to academic settings, with no successful clinical translation reported. The unknown differentiation process in vivo, potential tumorigenicity, and epigenetic abnormality of transplanted cells are preventing their clinical application. In this review, we give an overview on preclinical development of iPSC-derived myogenic cell transplantation therapies including processes related to iPSC-derived myogenic cells such as differentiation, scaling-up, delivery, and cGMP compliance. And we discuss the potential challenges of each step of clinical translation. Additionally, preclinical model systems for testing myogenic cells intended for clinical applications are described.
Collapse
Affiliation(s)
- Congshan Sun
- Vita Therapeutics, Baltimore, MD 21043, USA (M.R.)
| | - Carlo Serra
- Department of Neurology, Baylor College of Medicine, Houston, TX 77030, USA
| | | | | | - Mahendra Rao
- Vita Therapeutics, Baltimore, MD 21043, USA (M.R.)
| |
Collapse
|
20
|
Chugh S, Bahal RK, Dhiman R, Singh R. Antigen identification strategies and preclinical evaluation models for advancing tuberculosis vaccine development. NPJ Vaccines 2024; 9:57. [PMID: 38461350 PMCID: PMC10924964 DOI: 10.1038/s41541-024-00834-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 02/05/2024] [Indexed: 03/11/2024] Open
Abstract
In its myriad devastating forms, Tuberculosis (TB) has existed for centuries, and humanity is still affected by it. Mycobacterium tuberculosis (M. tuberculosis), the causative agent of TB, was the foremost killer among infectious agents until the COVID-19 pandemic. One of the key healthcare strategies available to reduce the risk of TB is immunization with bacilli Calmette-Guerin (BCG). Although BCG has been widely used to protect against TB, reports show that BCG confers highly variable efficacy (0-80%) against adult pulmonary TB. Unwavering efforts have been made over the past 20 years to develop and evaluate new TB vaccine candidates. The failure of conventional preclinical animal models to fully recapitulate human response to TB, as also seen for the failure of MVA85A in clinical trials, signifies the need to develop better preclinical models for TB vaccine evaluation. In the present review article, we outline various approaches used to identify protective mycobacterial antigens and recent advancements in preclinical models for assessing the efficacy of candidate TB vaccines.
Collapse
Affiliation(s)
- Saurabh Chugh
- Centre for Tuberculosis Research, Tuberculosis Research Laboratory, Translational Health Science and Technology Institute, Faridabad, 121001, Haryana, India
| | - Ritika Kar Bahal
- Marshall Centre, School of Biomedical Sciences, University of Western Australia, Perth, Australia
| | - Rohan Dhiman
- Laboratory of Mycobacterial Immunology, Department of Life Science, National Institute of Technology, Rourkela, 769008, Odisha, India
| | - Ramandeep Singh
- Centre for Tuberculosis Research, Tuberculosis Research Laboratory, Translational Health Science and Technology Institute, Faridabad, 121001, Haryana, India.
| |
Collapse
|
21
|
Yang Y, Li J, Li D, Zhou W, Yan F, Wang W. Humanized mouse models: A valuable platform for preclinical evaluation of human cancer. Biotechnol Bioeng 2024; 121:835-852. [PMID: 38151887 DOI: 10.1002/bit.28618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 11/26/2023] [Indexed: 12/29/2023]
Abstract
Animal models are routinely employed to assess the treatments for human cancer. However, due to significant differences in genetic backgrounds, traditional animal models are unable to meet bioresearch needs. To overcome this restriction, researchers have generated and optimized immunodeficient mice, and then engrafted human genes, cells, tissues, or organs in mice so that the responses in the model mice could provide a more reliable reference for treatments. As a bridge connecting clinical application and basic research, humanized mice are increasingly used in the preclinical evaluation of cancer treatments, particularly after gene interleukin 2 receptor gamma mutant mice were generated. Human cancer models established in humanized mice support exploration of the mechanism of cancer occurrence and provide an efficient platform for drug screening. However, it is undeniable that the further application of humanized mice still faces multiple challenges. This review summarizes the construction approaches for humanized mice and their existing limitations. We also report the latest applications of humanized mice in preclinical evaluation for the treatment of cancer and point out directions for future optimization of these models.
Collapse
Affiliation(s)
- Yuening Yang
- State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Jiaqian Li
- State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Dan Li
- State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Weilin Zhou
- State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Feiyang Yan
- State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Wei Wang
- State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
22
|
Lundy J. A Humanized Patient-Derived Xenograft Model for Pancreatic Cancer. Methods Mol Biol 2024; 2806:91-100. [PMID: 38676798 DOI: 10.1007/978-1-0716-3858-3_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/29/2024]
Abstract
Pancreatic cancer is associated with a high mortality rate, and there are still very few effective treatment options. Patient-derived xenografts have proven to be invaluable preclinical disease models to study cancer biology and facilitate testing of novel therapeutics. However, the severely immune-deficient mice used to generate standard models lack any functional immune system, thereby limiting their utility as a tool to investigate the tumor-immune cell interface. This chapter will outline a method for establishment of "humanized" patient-derived xenografts, which are reconstituted with human immune cells to imitate the immune-rich microenvironment of pancreatic cancer.
Collapse
Affiliation(s)
- Joanne Lundy
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC, Australia.
- Department of Molecular Translational Science, School of Clinical Sciences, Monash University, Clayton, VIC, Australia.
- Department of Surgery, School of Clinical Sciences, Monash University, Clayton, VIC, Australia.
- Peninsula Clinical School, Central Clinical School, Faculty of Medicine Nursing and Health Sciences, Monash University, Clayton, VIC, Australia.
| |
Collapse
|
23
|
Weng T, Jenkins BJ, Saad MI. Patient-Derived Xenografts: A Valuable Preclinical Model for Drug Development and Biomarker Discovery. Methods Mol Biol 2024; 2806:19-30. [PMID: 38676793 DOI: 10.1007/978-1-0716-3858-3_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/29/2024]
Abstract
Patient-derived xenografts (PDXs), established by implanting patient tumor cells into immunodeficient mice, offer a platform for faithfully replicating human tumors. They closely mimic the histopathology, genomics, and drug sensitivity of patient tumors. This chapter highlights the versatile applications of PDXs, including studying tumor biology, metastasis, and chemoresistance, as well as their use in biomarker identification, drug screening, and personalized medicine. It also addresses challenges in using PDXs in cancer research, including variations in metastatic potential, lengthy establishment timelines, stromal changes, and limitations in immunocompromised models. Despite these challenges, PDXs remain invaluable tools guiding patient treatment and advancing preclinical drug development.
Collapse
Affiliation(s)
- Teresa Weng
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Molecular and Translational Sciences, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC, Australia
| | - Brendan J Jenkins
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Molecular and Translational Sciences, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC, Australia
- South Australian immunoGENomics Cancer Institute (SAiGENCI), University of Adelaide, Adelaide, SA, Australia
| | - Mohamed I Saad
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC, Australia.
- Department of Molecular and Translational Sciences, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC, Australia.
- South Australian immunoGENomics Cancer Institute (SAiGENCI), University of Adelaide, Adelaide, SA, Australia.
| |
Collapse
|
24
|
Thompson J, Saad MI. Patient-Derived Xenografts: Historical Evolution, Immunocompromised Host Models, and Translational Significance. Methods Mol Biol 2024; 2806:1-8. [PMID: 38676791 DOI: 10.1007/978-1-0716-3858-3_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/29/2024]
Abstract
Patient-derived xenografts (PDXs) represent a critical advancement in preclinical cancer research, wherein human tumor samples are implanted into animal models for evaluation of therapeutic responses. PDXs have emerged as indispensable tools in translational cancer research, facilitating investigation into tumor microenvironments and personalized medicine. This chapter elucidates the historical evolution of PDXs, from early attempts in the eighteenth century to contemporary immunocompromised host models that enhance engraftment success.
Collapse
Affiliation(s)
- James Thompson
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Molecular and Translational Sciences, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC, Australia
| | - Mohamed I Saad
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC, Australia.
- Department of Molecular and Translational Sciences, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC, Australia.
- South Australian immunoGENomics Cancer Institute (SAiGENCI), University of Adelaide, Adelaide, SA, Australia.
| |
Collapse
|
25
|
Kaczmarek K, Więckiewicz J, Que I, Gałuszka-Bulaga A, Chan A, Siedlar M, Baran J. Human Soluble TRAIL Secreted by Modified Lactococcus lactis Bacteria Promotes Tumor Growth in the Orthotopic Mouse Model of Colorectal Cancer. Arch Immunol Ther Exp (Warsz) 2024; 72:aite-2024-0002. [PMID: 38299562 DOI: 10.2478/aite-2024-0002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 11/30/2023] [Indexed: 02/02/2024]
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) selectively induces apoptosis of sensitive cancer cells, including colorectal cancer (CRC). Due to its short biological half-life after intravenous administration and related clinical ineffectiveness, novel formulations of TRAIL need to be developed. Here we propose Lactococcus lactis bacteria as a vehicle for local delivery of human soluble TRAIL (hsTRAIL) in CRC. The use of common probiotics targeting guts as carriers for TRAIL could ensure its sustained release at the tumor site and extend the duration of its activity. We have already engineered hsTRAIL-secreting L.lactis bacteria and showed their effectiveness in elimination of human CRC cells in vitro and in vivo in a mouse subcutaneous model. Here, L.lactis(hsTRAIL+) were administered by gastric gavage to SCID mice with orthotopically developed HCT116 tumor in cecum, in monotherapy or in combination with metformin (MetF), already shown to enhance the hsTRAIL anti-tumor activity in subcutaneous CRC model. Oral administration of L.lactis(hsTRAIL+) resulted in significant progression of HCT116 tumors and shortening of the colon crypts. Secretion of hsTRAIL in the colon was accompanied by infiltration of the primary tumor with M2-macrophages, while MetF promoted transient colonization of the gut by L.lactis. Our study indicates that L.lactis bacteria after oral administration enable delivery of biologically active hsTRAIL to colon, however its potential therapeutic effect in CRC treatment is abolished by its pro-tumorigenic signalling, leading to the recruitment of M2-macrophages and tumor growth promotion.
Collapse
Affiliation(s)
- Katarzyna Kaczmarek
- Department of Clinical Immunology, Institute of Pediatrics, Jagiellonian University Medical College, Kraków, Poland
| | - Jerzy Więckiewicz
- Department of Clinical Immunology, Institute of Pediatrics, Jagiellonian University Medical College, Kraków, Poland
| | - Ivo Que
- Translational Nanobiomaterials and Imaging, Department of Radiology, Leiden University Medical Center, Albinusdreef 2, 2300 RC Leiden, The Netherlands
- Currently: Department of Radiology and Nuclear Medicine, Department of Molecular Genetics, Erasmus University Medical Center, Dr. Molewaterplein 40, 3015 GD Rotterdam, The Netherlands
| | - Adrianna Gałuszka-Bulaga
- Department of Clinical Immunology, Institute of Pediatrics, Jagiellonian University Medical College, Kraków, Poland
| | - Alan Chan
- Percuros B.V., Zernikedreef 8, 2333 CL Leiden, The Netherlands
| | - Maciej Siedlar
- Department of Clinical Immunology, Institute of Pediatrics, Jagiellonian University Medical College, Kraków, Poland
| | - Jarek Baran
- Department of Clinical Immunology, Institute of Pediatrics, Jagiellonian University Medical College, Kraków, Poland
| |
Collapse
|
26
|
Seon BK, Okazaki M, Duzen J, Matsuno F, Goey AKL, Maguire O. Identification of unique molecular heterogeneity of human CD79, the signaling component of the human B cell antigen receptor (BCR), and synergistic potentiation of the CD79-targeted therapy of B cell tumors by co-targeting of CD79a and CD79b. Leuk Res 2024; 136:107436. [PMID: 38232613 PMCID: PMC10906460 DOI: 10.1016/j.leukres.2024.107436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 01/02/2024] [Accepted: 01/04/2024] [Indexed: 01/19/2024]
Abstract
We identified unique molecular heterogeneity of CD79 of human B cell antigen receptor (BCR) that may open a new approach to the ongoing CD79b-targeted therapy of B cell tumors. The primary purpose of the present study is to gain new information valuable for the enhanced CD79-targeted therapy. The molecular heterogeneity of CD79 was identified by sequential immunoprecipitation of BCR by use of anti-CD79b monoclonal antibody (mAb) SN8 and anti-CD79a mAb SN8b. SN8 is the antibody component of polatuzumab vedotin, an anti-CD79b antibody drug conjugate, that has been widely used for therapy of diffuse large B-cell lymphoma (DLBCL). The sequential immunoprecipitation shows that anti-CD79b mAb will be able to react only with a subgroup of CD79 molecules while anti-CD79a mAb will react with another subgroup of CD79 molecules; CD79 is a disulfide-linked heterodimer of CD79a and CD79b. Therapeutic study of SCID mice bearing human B-cell tumor shows synergistic potentiation by co-targeting CD79b and CD79a. Furthermore, simultaneous targeting of PD-1 strongly potentiates CD79a/CD79b-targeted therapy of B cell tumors. Flow cytometry analyses of CD79a/CD79b on malignant B cells of patients may provide a method for selection of the candidate patients for the CD79a/CD79b dual targeting therapy.
Collapse
Affiliation(s)
- Ben K Seon
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA.
| | - Morihiro Okazaki
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Jill Duzen
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Fumihiko Matsuno
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Andrew K L Goey
- Bioanalytics, Metabolomics and Pharmacokinetics (BMPK) Shared Resource, and Department of Pharmacology and Therapeutics, Rpswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Orla Maguire
- Flow and Image Cytometry Shared Resource, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| |
Collapse
|
27
|
Wang X, Chen M, Hu L, Tan C, Li X, Xue P, Jiang Y, Bao P, Yu T, Li F, Xiao Y, Ran Q, Li Z, Chen L. Humanized mouse models for inherited thrombocytopenia studies. Platelets 2023; 34:2267676. [PMID: 37849076 DOI: 10.1080/09537104.2023.2267676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 10/03/2023] [Indexed: 10/19/2023]
Abstract
Inherited thrombocytopenia (IT) is a group of hereditary disorders characterized by a reduced platelet count as the main clinical manifestation, and often with abnormal platelet function, which can subsequently lead to impaired hemostasis. In the past decades, humanized mouse models (HMMs), that are mice engrafted with human cells or genes, have been widely used in different research areas including immunology, oncology, and virology. With advances of the development of immunodeficient mice, the engraftment, and reconstitution of functional human platelets in HMM permit studies of occurrence and development of platelet disorders including IT and treatment strategies. This article mainly reviews the development of humanized mice models, the construction methods, research status, and problems of using humanized mice for the in vivo study of human thrombopoiesis.
Collapse
Affiliation(s)
- Xiaojie Wang
- Laboratory of Radiation Biology, Department of Blood Transfusion, Laboratory Medicine Center, The Second Affiliated Hospital, Army Medical University, Chongqing, China
- Basic Research Innovation Center for Prevention and Treatment of Acute Radiation Syndrome, The Second Affiliated Hospital, Third Military Medical University, Chongqing, China
| | - Maoshan Chen
- Laboratory of Radiation Biology, Department of Blood Transfusion, Laboratory Medicine Center, The Second Affiliated Hospital, Army Medical University, Chongqing, China
- Basic Research Innovation Center for Prevention and Treatment of Acute Radiation Syndrome, The Second Affiliated Hospital, Third Military Medical University, Chongqing, China
- Laboratory of Precision Medicine, Laboratory Medicine Center, The Second Affiliated Hospital, Third Military Medical University, Chongqing, China
| | - Lanyue Hu
- Laboratory of Radiation Biology, Department of Blood Transfusion, Laboratory Medicine Center, The Second Affiliated Hospital, Army Medical University, Chongqing, China
- Basic Research Innovation Center for Prevention and Treatment of Acute Radiation Syndrome, The Second Affiliated Hospital, Third Military Medical University, Chongqing, China
| | - Chengning Tan
- Laboratory of Radiation Biology, Department of Blood Transfusion, Laboratory Medicine Center, The Second Affiliated Hospital, Army Medical University, Chongqing, China
- Basic Research Innovation Center for Prevention and Treatment of Acute Radiation Syndrome, The Second Affiliated Hospital, Third Military Medical University, Chongqing, China
| | - Xiaoliang Li
- Laboratory of Radiation Biology, Department of Blood Transfusion, Laboratory Medicine Center, The Second Affiliated Hospital, Army Medical University, Chongqing, China
- Basic Research Innovation Center for Prevention and Treatment of Acute Radiation Syndrome, The Second Affiliated Hospital, Third Military Medical University, Chongqing, China
| | - Peipei Xue
- Laboratory of Radiation Biology, Department of Blood Transfusion, Laboratory Medicine Center, The Second Affiliated Hospital, Army Medical University, Chongqing, China
- Basic Research Innovation Center for Prevention and Treatment of Acute Radiation Syndrome, The Second Affiliated Hospital, Third Military Medical University, Chongqing, China
| | - Yangzhou Jiang
- Laboratory of Radiation Biology, Department of Blood Transfusion, Laboratory Medicine Center, The Second Affiliated Hospital, Army Medical University, Chongqing, China
| | - Peipei Bao
- Laboratory of Radiation Biology, Department of Blood Transfusion, Laboratory Medicine Center, The Second Affiliated Hospital, Army Medical University, Chongqing, China
| | - Teng Yu
- Laboratory of Radiation Biology, Department of Blood Transfusion, Laboratory Medicine Center, The Second Affiliated Hospital, Army Medical University, Chongqing, China
| | - Fengjie Li
- Laboratory of Radiation Biology, Department of Blood Transfusion, Laboratory Medicine Center, The Second Affiliated Hospital, Army Medical University, Chongqing, China
- Basic Research Innovation Center for Prevention and Treatment of Acute Radiation Syndrome, The Second Affiliated Hospital, Third Military Medical University, Chongqing, China
| | - Yanni Xiao
- Laboratory of Radiation Biology, Department of Blood Transfusion, Laboratory Medicine Center, The Second Affiliated Hospital, Army Medical University, Chongqing, China
- Basic Research Innovation Center for Prevention and Treatment of Acute Radiation Syndrome, The Second Affiliated Hospital, Third Military Medical University, Chongqing, China
| | - Qian Ran
- Laboratory of Radiation Biology, Department of Blood Transfusion, Laboratory Medicine Center, The Second Affiliated Hospital, Army Medical University, Chongqing, China
- Basic Research Innovation Center for Prevention and Treatment of Acute Radiation Syndrome, The Second Affiliated Hospital, Third Military Medical University, Chongqing, China
| | - Zhongjun Li
- Laboratory of Radiation Biology, Department of Blood Transfusion, Laboratory Medicine Center, The Second Affiliated Hospital, Army Medical University, Chongqing, China
- Basic Research Innovation Center for Prevention and Treatment of Acute Radiation Syndrome, The Second Affiliated Hospital, Third Military Medical University, Chongqing, China
- Laboratory of Precision Medicine, Laboratory Medicine Center, The Second Affiliated Hospital, Third Military Medical University, Chongqing, China
| | - Li Chen
- Laboratory of Radiation Biology, Department of Blood Transfusion, Laboratory Medicine Center, The Second Affiliated Hospital, Army Medical University, Chongqing, China
- Basic Research Innovation Center for Prevention and Treatment of Acute Radiation Syndrome, The Second Affiliated Hospital, Third Military Medical University, Chongqing, China
- Laboratory of Precision Medicine, Laboratory Medicine Center, The Second Affiliated Hospital, Third Military Medical University, Chongqing, China
| |
Collapse
|
28
|
Takagi Y, Sudo K, Yamaguchi S, Urata S, Ohno T, Hirose S, Matsumoto K, Kuramoto T, Serikawa T, Yasuda J, Ikutani M, Nakae S. Characterization of novel, severely immunodeficient Prkdc Δex57/Δex57 mice. Biochem Biophys Res Commun 2023; 678:193-199. [PMID: 37651888 DOI: 10.1016/j.bbrc.2023.08.055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 08/25/2023] [Indexed: 09/02/2023]
Abstract
Severely immunodeficient mice are useful for understanding the pathogenesis of certain tumors and for developing therapeutic agents for such tumors. In addition, engraftment of these mice with human hematopoietic cells can yield information that helps us understand the in vivo molecular mechanisms underlying actual human viral infections. In our present research, we discovered a novel, severely immunodeficient strain of mice having a mutation in exon 57 of the Prkdc gene (PrkdcΔex57/Δex57) in an inbred colony of B10.S/SgSlc mice. Those PrkdcΔex57/Δex57 mice showed thymic hypoplasia and lack of mature T cells and B cells in peripheral lymphoid tissues, resulting in very low levels of production of serum immunoglobulins. In addition, those mice were highly susceptible to influenza viruses due to the lack of acquired immune cells. On the other hand, since they had sufficient numbers of NK cells, they rejected tumor transplants, similarly to Prkdc+/+ mice. Next, we generated Foxn1nu/nuPrkdcΔex57/Δex57Il2rg-/- (NPG) mice on the BALB/cSlc background, which lack all lymphocytes such as T cells, B cells and innate lymphoid cells, including NK cells. As expected, these mice were able to undergo engraftment of human tumor cell lines. These findings suggest that PrkdcΔex57/Δex57 mice will be useful as a novel model of immunodeficiency, while NPG mice will be useful for xenografting of various malignancies.
Collapse
Affiliation(s)
| | - Katsuko Sudo
- Preclinical Research Center, Tokyo Medical University, Tokyo, 160-8402, Japan
| | - Sachiko Yamaguchi
- Laboratory of Systems Biology, Center for Experimental Medicine and Systems Biology, Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639, Japan; Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, 739-8528, Japan
| | - Shuzo Urata
- National Research Center for the Control and Prevention of Infectious Diseases, Nagasaki University, Nagasaki, 852-8523, Japan
| | - Tatsukuni Ohno
- Oral Health Science Center, Tokyo Dental College, Tokyo, 101-0061, Japan
| | - Sachiko Hirose
- Toin Human Science and Technology Center, Department of Biomedical Engineering, Toin University of Yokohama, Yokohama, 225-8503, Japan
| | - Kiyoshi Matsumoto
- Division of Animal Research, Research Center for Advanced Science and Technology, Shinshu University, Nagano, 390-8621, Japan
| | - Takashi Kuramoto
- Department of Animal Science, Faculty of Agriculture, Tokyo University of Agriculture, Kanagawa, 243-0034, Japan
| | - Tadao Serikawa
- Institute of Laboratory Animals, Graduate School of Medicine, Kyoto University, Kyoto, 606-8501, Japan
| | - Jiro Yasuda
- National Research Center for the Control and Prevention of Infectious Diseases, Nagasaki University, Nagasaki, 852-8523, Japan
| | - Masashi Ikutani
- Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, 739-8528, Japan.
| | - Susumu Nakae
- Laboratory of Systems Biology, Center for Experimental Medicine and Systems Biology, Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639, Japan; Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, 739-8528, Japan.
| |
Collapse
|
29
|
Hartmann KP, van Gogh M, Freitag PC, Kast F, Nagy-Davidescu G, Borsig L, Plückthun A. FAP-retargeted Ad5 enables in vivo gene delivery to stromal cells in the tumor microenvironment. Mol Ther 2023; 31:2914-2928. [PMID: 37641405 PMCID: PMC10556229 DOI: 10.1016/j.ymthe.2023.08.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 06/19/2023] [Accepted: 08/24/2023] [Indexed: 08/31/2023] Open
Abstract
Fibroblast activation protein (FAP) is a cell surface serine protease that is highly expressed on reactive stromal fibroblasts, such as cancer-associated fibroblasts (CAFs), and generally absent in healthy adult tissues. FAP expression in the tumor stroma has been detected in more than 90% of all carcinomas, rendering CAFs excellent target cells for a tumor site-specific adenoviral delivery of cancer therapeutics. Here, we present a tropism-modified human adenovirus 5 (Ad5) vector that targets FAP through trivalent, designed ankyrin repeat protein-based retargeting adapters. We describe the development and validation of these adapters via cell-based screening assays and demonstrate adapter-mediated Ad5 retargeting to FAP+ fibroblasts in vitro and in vivo. We further show efficient in vivo delivery and in situ production of a therapeutic payload by CAFs in the tumor microenvironment (TME), resulting in attenuated tumor growth. We thus propose using our FAP-Ad5 vector to convert CAFs into a "biofactory," secreting encoded cancer therapeutics into the TME to enable a safe and effective cancer treatment.
Collapse
Affiliation(s)
- K Patricia Hartmann
- Department of Biochemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Merel van Gogh
- Department of Physiology, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Patrick C Freitag
- Department of Biochemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Florian Kast
- Department of Biochemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Gabriela Nagy-Davidescu
- Department of Biochemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Lubor Borsig
- Department of Physiology, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Andreas Plückthun
- Department of Biochemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland.
| |
Collapse
|
30
|
Pelosi AC, Scariot PPM, Garbuio ALP, Kraemer MB, Priolli DG, Masselli Dos Reis IG, Messias LHD. A systematic review of exercise protocols applied to athymic mice in tumor-related experiments. Appl Physiol Nutr Metab 2023; 48:719-729. [PMID: 37384946 DOI: 10.1139/apnm-2023-0027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/01/2023]
Abstract
Athymic mice are unable to produce T-cells and are then characterized as immunodeficient. This characteristic makes these animals ideal for tumor biology and xenograft research. New non-pharmacological therapeutics are required owing to the exponential increase in global oncology costs over the last 10 years and the high cancer mortality rate. In this sense, physical exercise is regarded as a relevant component of cancer treatment. However, the scientific community lacks information regarding the effect of manipulating training variables on cancer in humans, and experiments with athymic mice. Therefore, this systematic review aimed to address the exercise protocols used in tumor-related experiments using athymic mice. The PubMed, Web of Science, and Scopus databases were searched without restrictions on published data. A combination of key terms such as athymic mice, nude mice, physical activity, physical exercise, and training was used. The database search retrieved 852 studies (PubMed, 245; Web of Science, 390; and Scopus, 217). After title, abstract, and full-text screening, 10 articles were eligible. Based on the included studies, this report highlights the considerable divergences in the training variables adopted for this animal model. No studies have reported the determination of a physiological marker for intensity individualization. Future studies are recommended to explore whether invasive procedures can result in pathogenic infections in athymic mice. Moreover, time-consuming tests cannot be applied to experiments with specific characteristics such as tumor implantation. In summary, non-invasive, low-cost, and time-saving approaches can suppress these limitations and improve the welfare of these animals during experiments.
Collapse
Affiliation(s)
- Andrea Corazzi Pelosi
- Research Group on Technology Applied to Exercise Physiology (GTAFE), Laboratory of Multidisciplinary Research, São Francisco University, Bragança Paulista, Brazil
| | | | - Ana Luíza Paula Garbuio
- Research Group on Technology Applied to Exercise Physiology (GTAFE), Laboratory of Multidisciplinary Research, São Francisco University, Bragança Paulista, Brazil
| | - Maurício Beitia Kraemer
- Research Group on Technology Applied to Exercise Physiology (GTAFE), Laboratory of Multidisciplinary Research, São Francisco University, Bragança Paulista, Brazil
| | - Denise Gonçalves Priolli
- Coloproctology service of the Federal University of São Paulo, São Paulo and Faculty of Health Sciences Pitágoras de Codó, Codó, Brazil
| | - Ivan Gustavo Masselli Dos Reis
- Research Group on Technology Applied to Exercise Physiology (GTAFE), Laboratory of Multidisciplinary Research, São Francisco University, Bragança Paulista, Brazil
| | - Leonardo Henrique Dalcheco Messias
- Research Group on Technology Applied to Exercise Physiology (GTAFE), Laboratory of Multidisciplinary Research, São Francisco University, Bragança Paulista, Brazil
| |
Collapse
|
31
|
Schmid-Siegert E, Qin M, Tian H, Arpat B, Chen B, Xenarios I. Reference genomes for BALB/c Nude and NOD/SCID mouse models. G3 (BETHESDA, MD.) 2023; 13:jkad188. [PMID: 37594081 PMCID: PMC10542179 DOI: 10.1093/g3journal/jkad188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 08/01/2023] [Indexed: 08/19/2023]
Abstract
Mouse xenograft models play a vital role in tumor studies for research as well as for screening of drugs for the pharmaceutical industry. In particular, models with compromised immunity are favorable to increase the success of transplantation, such as, e.g. NOD/SCID and BALB/c Nude strains. The genomic sequence and alterations of many of these models still remain elusive and might hamper a model's further optimization or proper adapted usage. This can be in respect to treatments (e.g. NOD/SCID sensitivity to radiation), experiments or analysis of derived sequencing data of such models. Here we present the genome assemblies for the NOD/SCID and BALB/c Nude strains to overcome this short-coming for the future and improve our understanding of these models in the process. We highlight as well first insights into observed genomic differences for these models compared to the C57BL/6 reference genome. Genome assemblies for both are close to full-chromosome representations and provided with liftover annotations from the GRCm39 reference genome.
Collapse
Affiliation(s)
- Emanuel Schmid-Siegert
- JSR Life Sciences, NGS-AI CH DivisionRoute de la Corniche 3, 1066 Epalinges, Switzerland
| | - Mengting Qin
- JSR Life Sciences, NGS-AI CN Division, Industrial Park, Suzhou, Jiangsu 215000, P.R. China
| | - Huan Tian
- JSR Life Sciences, NGS-AI CN Division, Industrial Park, Suzhou, Jiangsu 215000, P.R. China
| | - Bulak Arpat
- JSR Life Sciences, NGS-AI CH DivisionRoute de la Corniche 3, 1066 Epalinges, Switzerland
| | - Bonnie Chen
- JSR Life Sciences, NGS-AI CN Division, Industrial Park, Suzhou, Jiangsu 215000, P.R. China
| | - Ioannis Xenarios
- JSR Life Sciences, NGS-AI CH DivisionRoute de la Corniche 3, 1066 Epalinges, Switzerland
| |
Collapse
|
32
|
Candelaria PV, Nava M, Daniels-Wells TR, Penichet ML. A Fully Human IgE Specific for CD38 as a Potential Therapy for Multiple Myeloma. Cancers (Basel) 2023; 15:4533. [PMID: 37760502 PMCID: PMC10526502 DOI: 10.3390/cancers15184533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 08/30/2023] [Accepted: 08/30/2023] [Indexed: 09/29/2023] Open
Abstract
Multiple myeloma (MM) is an incurable malignancy of plasma cells and the second most common hematologic malignancy in the United States. Although antibodies in clinical cancer therapy are generally of the IgG class, antibodies of the IgE class have attractive properties as cancer therapeutics, such as their high affinity for Fc receptors (FcεRs), the low serum levels of endogenous IgE allowing for less competition for FcR occupancy, and the lack of inhibitory FcRs. Importantly, the FcεRs are expressed on immune cells that elicit antibody-dependent cell-mediated cytotoxicity (ADCC), antibody-dependent cell-mediated phagocytosis (ADCP), and/or antigen presentation such as mast cells, eosinophils, macrophages, and dendritic cells. We now report the development of a fully human IgE targeting human CD38 as a potential MM therapy. We targeted CD38 given its high and uniform expression on MM cells. The novel anti-CD38 IgE, expressed in mammalian cells, is properly assembled and secreted, exhibits the correct molecular weight, binds antigen and the high affinity FcεRI, and induces degranulation of FcεRI expressing cells in vitro and also in vivo in transgenic BALB/c mice expressing human FcεRIα. Moreover, the anti-CD38 IgE induces ADCC and ADCP mediated by monocytes/macrophages against human MM cells (MM.1S). Importantly, the anti-CD38 IgE also prolongs survival in a preclinical disseminated xenograft mouse model using SCID-Beige mice and human MM.1S cells when administered with human peripheral blood mononuclear cells (PBMCs) as a source of monocyte effector cells. Our results suggest that anti-CD38 IgE may be effective in humans bearing MM and other malignancies expressing CD38.
Collapse
Affiliation(s)
- Pierre V. Candelaria
- Division of Surgical Oncology, Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA
| | - Miguel Nava
- Division of Surgical Oncology, Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA
| | - Tracy R. Daniels-Wells
- Division of Surgical Oncology, Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA
| | - Manuel L. Penichet
- Division of Surgical Oncology, Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA
- UCLA AIDS Institute, Los Angeles, CA 90095, USA
- UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA 90095, USA
- The Molecular Biology Institute, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA
| |
Collapse
|
33
|
Zheng HC, Xue H, Yun WJ. An overview of mouse models of hepatocellular carcinoma. Infect Agent Cancer 2023; 18:49. [PMID: 37670307 PMCID: PMC10481604 DOI: 10.1186/s13027-023-00524-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 08/21/2023] [Indexed: 09/07/2023] Open
Abstract
Hepatocellular carcinoma (HCC) has become a severe burden on global health due to its high morbidity and mortality rates. However, effective treatments for HCC are limited. The lack of suitable preclinical models may contribute to a major failure of drug development for HCC. Here, we overview several well-established mouse models of HCC, including genetically engineered mice, chemically-induced models, implantation models, and humanized mice. Immunotherapy studies of HCC have been a hot topic. Therefore, we will introduce the application of mouse models of HCC in immunotherapy. This is followed by a discussion of some other models of HCC-related liver diseases, including non-alcoholic fatty liver disease (NAFLD), hepatitis B and C virus infection, and liver fibrosis and cirrhosis. Together these provide researchers with a current overview of the mouse models of HCC and assist in the application of appropriate models for their research.
Collapse
Affiliation(s)
- Hua-Chuan Zheng
- Department of Oncology and Central Laboratory, The Affiliated Hospital of Chengde Medical University, Chengde, 067000, China.
| | - Hang Xue
- Department of Oncology and Central Laboratory, The Affiliated Hospital of Chengde Medical University, Chengde, 067000, China
| | - Wen-Jing Yun
- Department of Oncology and Central Laboratory, The Affiliated Hospital of Chengde Medical University, Chengde, 067000, China
| |
Collapse
|
34
|
Mahmoudian RA, Farshchian M, Golyan FF, Mahmoudian P, Alasti A, Moghimi V, Maftooh M, Khazaei M, Hassanian SM, Ferns GA, Mahaki H, Shahidsales S, Avan A. Preclinical tumor mouse models for studying esophageal cancer. Crit Rev Oncol Hematol 2023; 189:104068. [PMID: 37468084 DOI: 10.1016/j.critrevonc.2023.104068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 07/13/2023] [Accepted: 07/14/2023] [Indexed: 07/21/2023] Open
Abstract
Preclinical models are extensively employed in cancer research because they can be manipulated in terms of their environment, genome, molecular biology, organ systems, and physical activity to mimic human behavior and conditions. The progress made in in vivo cancer research has resulted in significant advancements, enabling the creation of spontaneous, metastatic, and humanized mouse models. Most recently, the remarkable and extensive developments in genetic engineering, particularly the utilization of CRISPR/Cas9, transposable elements, epigenome modifications, and liquid biopsies, have further facilitated the design and development of numerous mouse models for studying cancer. In this review, we have elucidated the production and usage of current mouse models, such as xenografts, chemical-induced models, and genetically engineered mouse models (GEMMs), for studying esophageal cancer. Additionally, we have briefly discussed various gene-editing tools that could potentially be employed in the future to create mouse models specifically for esophageal cancer research.
Collapse
Affiliation(s)
- Reihaneh Alsadat Mahmoudian
- Cancer Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Moein Farshchian
- Division of Oncology, Laboratory of Cellular Therapy, Department of Medical and Surgical Sciences for Children and Adults, University Hospital of Modena and Reggio Emilia, Modena, Italy
| | - Fatemeh Fardi Golyan
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Parvaneh Mahmoudian
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali Alasti
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Vahid Moghimi
- Department of Biology, Faculty of Science, Hakim Sabzevari University, Sabzevar, Iran
| | - Mina Maftooh
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Majid Khazaei
- Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Mahdi Hassanian
- Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gordon A Ferns
- Brighton & Sussex Medical School, Department of Medical Education, Falmer, Brighton, Sussex BN1 9PH, UK
| | - Hanie Mahaki
- Vascular & Endovascular Surgery Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Amir Avan
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; College of Medicine, University of Warith Al-Anbiyaa, Karbala, Iraq; Faculty of Health, School of Biomedical Sciences, Queensland University of Technology, Brisbane, Australia.
| |
Collapse
|
35
|
Olkowski C, Fernandes B, Griffiths GL, Lin F, Choyke PL. Preclinical Imaging of Prostate Cancer. Semin Nucl Med 2023; 53:644-662. [PMID: 36882335 PMCID: PMC10440231 DOI: 10.1053/j.semnuclmed.2023.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 02/01/2023] [Indexed: 03/07/2023]
Abstract
Prostate cancer remains a major cause of mortality and morbidity, affecting millions of men, with a large percentage expected to develop the disease as they reach advanced ages. Treatment and management advances have been dramatic over the past 50 years or so, and one aspect of these improvements is reflected in the multiple advances in diagnostic imaging techniques. Much attention has been focused on molecular imaging techniques that offer high sensitivity and specificity and can now more accurately assess disease status and detect recurrence earlier. During development of molecular imaging probes, single-photon emission computed tomography (SPECT) and positron emission tomography (PET) must be evaluated in preclinical models of the disease. If such agents are to be translated to the clinic, where patients undergoing these imaging modalities are injected with a molecular imaging probe, these agents must first be approved by the FDA and other regulatory agencies prior to their adoption in clinical practice. Scientists have worked assiduously to develop preclinical models of prostate cancer that are relevant to the human disease to enable testing of these probes and related targeted drugs. Challenges in developing reproducible and robust models of human disease in animals are beset with practical issues such as the lack of natural occurrence of prostate cancer in mature male animals, the difficulty of initiating disease in immune-competent animals and the sheer size differences between humans and conveniently smaller animals such as rodents. Thus, compromises in what is ideal and what can be achieved have had to be made. The workhorse of preclinical animal models has been, and remains, the investigation of human xenograft tumor models in athymic immunocompromised mice. Later models have used other immunocompromised models as they have been found and developed, including the use of directly derived patient tumor tissues, completely immunocompromised mice, orthotopic methods for inducing prostate cancer within the mouse prostate itself and metastatic models of advanced disease. These models have been developed in close parallel with advances in imaging agent chemistries, radionuclide developments, computer electronics advances, radiometric dosimetry, biotechnologies, organoid technologies, advances in in vitro diagnostics, and overall deeper understandings of disease initiation, development, immunology, and genetics. The combination of molecular models of prostatic disease with radiometric-based studies in small animals will always remain spatially limited due to the inherent resolution sensitivity limits of PET and SPECT decay processes, fundamentally set at around a 0.5 cm resolution limit. Nevertheless, it is central to researcher's efforts and to successful clinical translation that the best animal models are adopted, accepted, and scientifically verified as part of this truly interdisciplinary approach to addressing this important disease.
Collapse
Affiliation(s)
- Colleen Olkowski
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, Bethesda MD
| | - Bruna Fernandes
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, Bethesda MD
| | - Gary L Griffiths
- Clinical Research Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD
| | - Frank Lin
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, Bethesda MD
| | - Peter L Choyke
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, Bethesda MD.
| |
Collapse
|
36
|
Lang Y, Lyu Y, Tan Y, Hu Z. Progress in construction of mouse models to investigate the pathogenesis and immune therapy of human hematological malignancy. Front Immunol 2023; 14:1195194. [PMID: 37646021 PMCID: PMC10461088 DOI: 10.3389/fimmu.2023.1195194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 07/27/2023] [Indexed: 09/01/2023] Open
Abstract
Hematological malignancy is a disease arisen by complicate reasons that seriously endangers human health. The research on its pathogenesis and therapies depends on the usage of animal models. Conventional animal model cannot faithfully mirror some characteristics of human features due to the evolutionary divergence, whereas the mouse models hosting human hematological malignancy are more and more applied in basic as well as translational investigations in recent years. According to the construction methods, they can be divided into different types (e.g. cell-derived xenograft (CDX) and patient-derived xenograft model (PDX) model) that have diverse characteristics and application values. In addition, a variety of strategies have been developed to improve human hematological malignant cell engraftment and differentiation in vivo. Moreover, the humanized mouse model with both functional human immune system and autologous human hematological malignancy provides a unique tool for the evaluation of the efficacy of novel immunotherapeutic drugs/approaches. Herein, we first review the evolution of the mouse model of human hematological malignancy; Then, we analyze the characteristics of different types of models and summarize the ways to improve the models; Finally, the way and value of humanized mouse model of human immune system in the immunotherapy of human hematological malignancy are discussed.
Collapse
Affiliation(s)
- Yue Lang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, China
- Department of Dermatology, The First Hospital, Jilin University, Changchun, China
| | - Yanan Lyu
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, China
| | - Yehui Tan
- Department of Hematology, The First Hospital, Jilin University, Changchun, China
| | - Zheng Hu
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, China
| |
Collapse
|
37
|
Sutcu HH, Montagne B, Ricchetti M. DNA-PKcs regulates myogenesis in an Akt-dependent manner independent of induced DNA damage. Cell Death Differ 2023; 30:1900-1915. [PMID: 37400716 PMCID: PMC10406879 DOI: 10.1038/s41418-023-01177-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 04/26/2023] [Accepted: 05/03/2023] [Indexed: 07/05/2023] Open
Abstract
Skeletal muscle regeneration relies on muscle stem (satellite) cells. We previously demonstrated that satellite cells efficiently and accurately repair radiation-induced DNA double-strand breaks (DSBs) via the DNA-dependent kinase DNA-PKcs. We show here that DNA-PKcs affects myogenesis independently of its role in DSB repair. Consequently, this process does not require the accumulation of DSBs and it is also independent of caspase-induced DNA damage. We report that in myogenic cells DNA-PKcs is essential for the expression of the differentiation factor Myogenin in an Akt2-dependent manner. DNA-PKcs interacts with the p300-containing complex that activates Myogenin transcription. We show also that SCID mice that are deficient in DNA-PKcs, and are used for transplantation and muscle regeneration studies, display altered myofiber composition and delayed myogenesis upon injury. These defects are exacerbated after repeated injury/regeneration events resulting in reduced muscle size. We thus identify a novel, caspase-independent, regulation of myogenic differentiation, and define a differentiation phase that does not involve the DNA damage/repair process.
Collapse
Affiliation(s)
- Haser Hasan Sutcu
- Institut Pasteur, Team Stability of Nuclear & Mitochondrial DNA, Department of Developmental and Stem Cell Biology, CNRS UMR3738, 75015, Paris, France
- Université Pierre et Marie Curie (Sorbonne Universities, ED515), Paris, France
- Institut de Radioprotection et de Sûrété Nucléaire (IRSN), Radiobiology of Accidental Exposure Laboratory (PSE-SANTE/SERAMED/LRAcc), B.P. 17, 92262 Fontenay-aux-Roses, Cedex, France
| | - Benjamin Montagne
- Institut Pasteur, Team Stability of Nuclear & Mitochondrial DNA, Department of Developmental and Stem Cell Biology, CNRS UMR3738, 75015, Paris, France
- Institut Pasteur, Molecular Mechanisms of Pathological and Physiological Ageing, Department of Developmental and Stem Cell Biology, Paris, France
| | - Miria Ricchetti
- Institut Pasteur, Team Stability of Nuclear & Mitochondrial DNA, Department of Developmental and Stem Cell Biology, CNRS UMR3738, 75015, Paris, France.
- Institut Pasteur, Molecular Mechanisms of Pathological and Physiological Ageing, Department of Developmental and Stem Cell Biology, Paris, France.
| |
Collapse
|
38
|
Benedetti F, Silvestri G, Saadat S, Denaro F, Latinovic OS, Davis H, Williams S, Bryant J, Ippodrino R, Rathinam CV, Gallo RC, Zella D. Mycoplasma DnaK increases DNA copy number variants in vivo. Proc Natl Acad Sci U S A 2023; 120:e2219897120. [PMID: 37459550 PMCID: PMC10372619 DOI: 10.1073/pnas.2219897120] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 06/14/2023] [Indexed: 07/20/2023] Open
Abstract
The human microbiota affects critical cellular functions, although the responsible mechanism(s) is still poorly understood. In this regard, we previously showed that Mycoplasma fermentans DnaK, an HSP70 chaperone protein, hampers the activity of important cellular proteins responsible for DNA integrity. Here, we describe a novel DnaK knock-in mouse model generated in our laboratory to study the effect of M. fermentans DnaK expression in vivo. By using an array-based comparative genomic hybridization assay, we demonstrate that exposure to DnaK was associated with a higher number of DNA copy number variants (CNVs) indicative of unbalanced chromosomal alterations, together with reduced fertility and a high rate of fetal abnormalities. Consistent with their implication in genetic disorders, one of these CNVs caused a homozygous Grid2 deletion, resulting in an aberrant ataxic phenotype that recapitulates the extensive biallelic deletion in the Grid2 gene classified in humans as autosomal recessive spinocerebellar ataxia 18. Our data highlight a connection between components of the human urogenital tract microbiota, namely Mycoplasmas, and genetic abnormalities in the form of DNA CNVs, with obvious relevant medical, diagnostic, and therapeutic implications.
Collapse
Affiliation(s)
- Francesca Benedetti
- Institute of Human Virology and Global Virus Network Center, University of Maryland School of Medicine, Baltimore, MD21201
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD21201
| | - Giovannino Silvestri
- Institute of Human Virology and Global Virus Network Center, University of Maryland School of Medicine, Baltimore, MD21201
- Department of Medicine, School of Medicine, University of Maryland School of Medicine, Baltimore, MD21201
| | - Saman Saadat
- Institute of Human Virology and Global Virus Network Center, University of Maryland School of Medicine, Baltimore, MD21201
| | - Frank Denaro
- Department of Biology, Morgan State University, Baltimore, MD21251
| | - Olga S. Latinovic
- Institute of Human Virology and Global Virus Network Center, University of Maryland School of Medicine, Baltimore, MD21201
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD21201
| | - Harry Davis
- Institute of Human Virology and Global Virus Network Center, University of Maryland School of Medicine, Baltimore, MD21201
| | - Sumiko Williams
- Institute of Human Virology and Global Virus Network Center, University of Maryland School of Medicine, Baltimore, MD21201
| | - Joseph Bryant
- Institute of Human Virology and Global Virus Network Center, University of Maryland School of Medicine, Baltimore, MD21201
| | | | - Chozha V. Rathinam
- Institute of Human Virology and Global Virus Network Center, University of Maryland School of Medicine, Baltimore, MD21201
- Department of Medicine, School of Medicine, University of Maryland School of Medicine, Baltimore, MD21201
| | - Robert C. Gallo
- Institute of Human Virology and Global Virus Network Center, University of Maryland School of Medicine, Baltimore, MD21201
- Department of Medicine, School of Medicine, University of Maryland School of Medicine, Baltimore, MD21201
| | - Davide Zella
- Institute of Human Virology and Global Virus Network Center, University of Maryland School of Medicine, Baltimore, MD21201
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD21201
| |
Collapse
|
39
|
Nakajima S, Okuma K. Mouse Models for HTLV-1 Infection and Adult T Cell Leukemia. Int J Mol Sci 2023; 24:11737. [PMID: 37511495 PMCID: PMC10380921 DOI: 10.3390/ijms241411737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 07/18/2023] [Accepted: 07/20/2023] [Indexed: 07/30/2023] Open
Abstract
Adult T cell leukemia (ATL) is an aggressive hematologic disease caused by human T cell leukemia virus type 1 (HTLV-1) infection. Various animal models of HTLV-1 infection/ATL have been established to elucidate the pathogenesis of ATL and develop appropriate treatments. For analyses employing murine models, transgenic and immunodeficient mice are used because of the low infectivity of HTLV-1 in mice. Each mouse model has different characteristics that must be considered before use for different HTLV-1 research purposes. HTLV-1 Tax and HBZ transgenic mice spontaneously develop tumors, and the roles of both Tax and HBZ in cell transformation and tumor growth have been established. Severely immunodeficient mice were able to be engrafted with ATL cell lines and have been used in preclinical studies of candidate molecules for the treatment of ATL. HTLV-1-infected humanized mice with an established human immune system are a suitable model to characterize cells in the early stages of HTLV-1 infection. This review outlines the characteristics of mouse models of HTLV-1 infection/ATL and describes progress made in elucidating the pathogenesis of ATL and developing related therapies using these mice.
Collapse
Affiliation(s)
- Shinsuke Nakajima
- Department of Microbiology, Faculty of Medicine, Kansai Medical University, Hirakata 573-1010, Osaka, Japan
| | - Kazu Okuma
- Department of Microbiology, Faculty of Medicine, Kansai Medical University, Hirakata 573-1010, Osaka, Japan
| |
Collapse
|
40
|
Kutle I, Dittrich A, Wirth D. Mouse Models for Human Herpesviruses. Pathogens 2023; 12:953. [PMID: 37513800 PMCID: PMC10384569 DOI: 10.3390/pathogens12070953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 07/13/2023] [Accepted: 07/15/2023] [Indexed: 07/30/2023] Open
Abstract
More than one hundred herpesviruses have been isolated from different species so far, with nine infecting humans. Infections with herpesviruses are characterized by life-long latency and represent a significant challenge for human health. To investigate the consequences of infections and identify novel treatment options, in vivo models are of particular relevance. The mouse has emerged as an economical small animal model to investigate herpesvirus infections. However, except for herpes simplex viruses (HSV-1, HSV-2), human herpesviruses cannot infect mice. Three natural herpesviruses have been identified in mice: mouse-derived cytomegalovirus (MCMV), mouse herpesvirus 68 (MHV-68), and mouse roseolovirus (MRV). These orthologues are broadly used to investigate herpesvirus infections within the natural host. In the last few decades, immunocompromised mouse models have been developed, allowing the functional engraftment of various human cells and tissues. These xenograft mice represent valuable model systems to investigate human-restricted viruses, making them particularly relevant for herpesvirus research. In this review, we describe the various mouse models used to study human herpesviruses, thereby highlighting their potential and limitations. Emphasis is laid on xenograft mouse models, covering the development and refinement of immune-compromised mice and their application in herpesvirus research.
Collapse
Affiliation(s)
- Ivana Kutle
- Research Group Model Systems for Infection, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany
| | - Anne Dittrich
- Research Group Model Systems for Infection, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
- InSCREENeX GmbH, Inhoffenstraße 7, 38124 Braunschweig, Germany
| | - Dagmar Wirth
- Research Group Model Systems for Infection, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany
| |
Collapse
|
41
|
Zhang C, Zaman LA, Poluektova LY, Gorantla S, Gendelman HE, Dash PK. Humanized Mice for Studies of HIV-1 Persistence and Elimination. Pathogens 2023; 12:879. [PMID: 37513726 PMCID: PMC10383313 DOI: 10.3390/pathogens12070879] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 06/06/2023] [Accepted: 06/23/2023] [Indexed: 07/30/2023] Open
Abstract
A major roadblock to achieving a cure for human immunodeficiency virus type one (HIV-1) is the persistence of latent viral infections in the cells and tissue compartments of an infected human host. Latent HIV-1 proviral DNA persists in resting memory CD4+ T cells and mononuclear phagocytes (MPs; macrophages, microglia, and dendritic cells). Tissue viral reservoirs of both cell types reside in the gut, lymph nodes, bone marrow, spleen, liver, kidney, skin, adipose tissue, reproductive organs, and brain. However, despite the identification of virus-susceptible cells, several limitations persist in identifying broad latent reservoirs in infected persons. The major limitations include their relatively low abundance, the precise identification of latently infected cells, and the lack of biomarkers for identifying latent cells. While primary MP and CD4+ T cells and transformed cell lines are used to interrogate mechanisms of HIV-1 persistence, they often fail to accurately reflect the host cells and tissue environments that carry latent infections. Given the host specificity of HIV-1, there are few animal models that replicate the natural course of viral infection with any precision. These needs underlie the importance of humanized mouse models as both valuable and cost-effective tools for studying viral latency and subsequently identifying means of eliminating it. In this review, we discuss the advantages and limitations of humanized mice for studies of viral persistence and latency with an eye toward using these models to test antiretroviral and excision therapeutics. The goals of this research are to use the models to address how and under which circumstances HIV-1 latency can be detected and eliminated. Targeting latent reservoirs for an ultimate HIV-1 cure is the task at hand.
Collapse
Affiliation(s)
| | | | | | | | | | - Prasanta K. Dash
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA (S.G.)
| |
Collapse
|
42
|
Parodi M, Astigiano S, Carrega P, Pietra G, Vitale C, Damele L, Grottoli M, Guevara Lopez MDLL, Ferracini R, Bertolini G, Roato I, Vitale M, Orecchia P. Murine models to study human NK cells in human solid tumors. Front Immunol 2023; 14:1209237. [PMID: 37388731 PMCID: PMC10301748 DOI: 10.3389/fimmu.2023.1209237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 06/02/2023] [Indexed: 07/01/2023] Open
Abstract
Since the first studies, the mouse models have provided crucial support for the most important discoveries on NK cells, on their development, function, and circulation within normal and tumor tissues. Murine tumor models were initially set to study murine NK cells, then, ever more sophisticated human-in-mice models have been developed to investigate the behavior of human NK cells and minimize the interferences from the murine environment. This review presents an overview of the models that have been used along time to study NK cells, focusing on the most popular NOG and NSG models, which work as recipients for the preparation of human-in-mice tumor models, the study of transferred human NK cells, and the evaluation of various enhancers of human NK cell function, including cytokines and chimeric molecules. Finally, an overview of the next generation humanized mice is also provided along with a discussion on how traditional and innovative in-vivo and in-vitro approaches could be integrated to optimize effective pre-clinical studies.
Collapse
Affiliation(s)
- Monica Parodi
- Unità Operativa UO Patologia e Immunologia Sperimentale, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Simonetta Astigiano
- Animal Facility, IRCCS Ospedale Policlinico San Martino Genova, Genova, Italy
| | - Paolo Carrega
- Laboratory of Immunology and Biotherapy, Department of Human Pathology, University of Messina, Messina, Italy
| | - Gabriella Pietra
- Unità Operativa UO Patologia e Immunologia Sperimentale, IRCCS Ospedale Policlinico San Martino, Genova, Italy
- Dipartimento di Medicina Sperimentale, Università di Genova, Genova, Italy
| | - Chiara Vitale
- Unità Operativa UO Patologia e Immunologia Sperimentale, IRCCS Ospedale Policlinico San Martino, Genova, Italy
- Dipartimento di Medicina Sperimentale, Università di Genova, Genova, Italy
| | - Laura Damele
- Unità Operativa UO Patologia e Immunologia Sperimentale, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Melania Grottoli
- Dipartimento di Medicina Sperimentale, Università di Genova, Genova, Italy
| | | | - Riccardo Ferracini
- Department of Surgical Sciences, Bone and Dental Bioengineering Laboratory, C.I.R Dental School, University of Turin, Turin, Italy
- Department of Surgical Sciences (DISC), University of Genoa, Genoa, Italy
| | - Giulia Bertolini
- “Epigenomics and Biomarkers of Solid Tumors”, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Ilaria Roato
- Department of Surgical Sciences, Bone and Dental Bioengineering Laboratory, C.I.R Dental School, University of Turin, Turin, Italy
| | - Massimo Vitale
- Unità Operativa UO Patologia e Immunologia Sperimentale, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Paola Orecchia
- Unità Operativa UO Patologia e Immunologia Sperimentale, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| |
Collapse
|
43
|
Chen A, Neuwirth I, Herndler-Brandstetter D. Modeling the Tumor Microenvironment and Cancer Immunotherapy in Next-Generation Humanized Mice. Cancers (Basel) 2023; 15:2989. [PMID: 37296949 PMCID: PMC10251926 DOI: 10.3390/cancers15112989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 05/10/2023] [Accepted: 05/28/2023] [Indexed: 06/12/2023] Open
Abstract
Cancer immunotherapy has brought significant clinical benefits to numerous patients with malignant disease. However, only a fraction of patients experiences complete and durable responses to currently available immunotherapies. This highlights the need for more effective immunotherapies, combination treatments and predictive biomarkers. The molecular properties of a tumor, intratumor heterogeneity and the tumor immune microenvironment decisively shape tumor evolution, metastasis and therapy resistance and are therefore key targets for precision cancer medicine. Humanized mice that support the engraftment of patient-derived tumors and recapitulate the human tumor immune microenvironment of patients represent a promising preclinical model to address fundamental questions in precision immuno-oncology and cancer immunotherapy. In this review, we provide an overview of next-generation humanized mouse models suitable for the establishment and study of patient-derived tumors. Furthermore, we discuss the opportunities and challenges of modeling the tumor immune microenvironment and testing a variety of immunotherapeutic approaches using human immune system mouse models.
Collapse
Affiliation(s)
| | | | - Dietmar Herndler-Brandstetter
- Center for Cancer Research, Medical University of Vienna and Comprehensive Cancer Center, 1090 Vienna, Austria; (A.C.); (I.N.)
| |
Collapse
|
44
|
Singhal SS, Garg R, Mohanty A, Garg P, Ramisetty SK, Mirzapoiazova T, Soldi R, Sharma S, Kulkarni P, Salgia R. Recent Advancement in Breast Cancer Research: Insights from Model Organisms-Mouse Models to Zebrafish. Cancers (Basel) 2023; 15:cancers15112961. [PMID: 37296923 DOI: 10.3390/cancers15112961] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 05/23/2023] [Accepted: 05/24/2023] [Indexed: 06/12/2023] Open
Abstract
Animal models have been utilized for decades to investigate the causes of human diseases and provide platforms for testing novel therapies. Indeed, breakthrough advances in genetically engineered mouse (GEM) models and xenograft transplantation technologies have dramatically benefited in elucidating the mechanisms underlying the pathogenesis of multiple diseases, including cancer. The currently available GEM models have been employed to assess specific genetic changes that underlay many features of carcinogenesis, including variations in tumor cell proliferation, apoptosis, invasion, metastasis, angiogenesis, and drug resistance. In addition, mice models render it easier to locate tumor biomarkers for the recognition, prognosis, and surveillance of cancer progression and recurrence. Furthermore, the patient-derived xenograft (PDX) model, which involves the direct surgical transfer of fresh human tumor samples to immunodeficient mice, has contributed significantly to advancing the field of drug discovery and therapeutics. Here, we provide a synopsis of mouse and zebrafish models used in cancer research as well as an interdisciplinary 'Team Medicine' approach that has not only accelerated our understanding of varied aspects of carcinogenesis but has also been instrumental in developing novel therapeutic strategies.
Collapse
Affiliation(s)
- Sharad S Singhal
- Department of Medical Oncology and Therapeutic Research, Beckman Research Institute, City of Hope Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| | - Rachana Garg
- Department of Surgery, Beckman Research Institute, City of Hope Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| | - Atish Mohanty
- Department of Medical Oncology and Therapeutic Research, Beckman Research Institute, City of Hope Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| | - Pankaj Garg
- Department of Chemistry, GLA University, Mathura 281406, Uttar Pradesh, India
| | - Sravani Keerthi Ramisetty
- Department of Medical Oncology and Therapeutic Research, Beckman Research Institute, City of Hope Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| | - Tamara Mirzapoiazova
- Department of Medical Oncology and Therapeutic Research, Beckman Research Institute, City of Hope Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| | - Raffaella Soldi
- Translational Genomics Research Institute, Phoenix, AZ 85338, USA
| | - Sunil Sharma
- Translational Genomics Research Institute, Phoenix, AZ 85338, USA
| | - Prakash Kulkarni
- Department of Medical Oncology and Therapeutic Research, Beckman Research Institute, City of Hope Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
- Department of Systems Biology, Beckman Research Institute, City of Hope Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| | - Ravi Salgia
- Department of Medical Oncology and Therapeutic Research, Beckman Research Institute, City of Hope Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| |
Collapse
|
45
|
Karnik I, Her Z, Neo SH, Liu WN, Chen Q. Emerging Preclinical Applications of Humanized Mouse Models in the Discovery and Validation of Novel Immunotherapeutics and Their Mechanisms of Action for Improved Cancer Treatment. Pharmaceutics 2023; 15:1600. [PMID: 37376049 DOI: 10.3390/pharmaceutics15061600] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 05/22/2023] [Accepted: 05/24/2023] [Indexed: 06/29/2023] Open
Abstract
Cancer therapeutics have undergone immense research over the past decade. While chemotherapies remain the mainstay treatments for many cancers, the advent of new molecular techniques has opened doors for more targeted modalities towards cancer cells. Although immune checkpoint inhibitors (ICIs) have demonstrated therapeutic efficacy in treating cancer, adverse side effects related to excessive inflammation are often reported. There is a lack of clinically relevant animal models to probe the human immune response towards ICI-based interventions. Humanized mouse models have emerged as valuable tools for pre-clinical research to evaluate the efficacy and safety of immunotherapy. This review focuses on the establishment of humanized mouse models, highlighting the challenges and recent advances in these models for targeted drug discovery and the validation of therapeutic strategies in cancer treatment. Furthermore, the potential of these models in the process of uncovering novel disease mechanisms is discussed.
Collapse
Affiliation(s)
- Isha Karnik
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Singapore
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore
| | - Zhisheng Her
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Singapore
| | - Shu Hui Neo
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Singapore
| | - Wai Nam Liu
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Singapore
| | - Qingfeng Chen
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Singapore
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, Immunos, Singapore 138648, Singapore
| |
Collapse
|
46
|
Imai T, Yoshida H, Machida Y, Kuramochi M, Ichikawa H, Kubo T, Takahashi M, Kato T. Alteration in molecular properties during establishment and passaging of endometrial carcinoma patient-derived xenografts. Sci Rep 2023; 13:8511. [PMID: 37231035 DOI: 10.1038/s41598-023-35703-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 05/22/2023] [Indexed: 05/27/2023] Open
Abstract
Patient-derived xenograft (PDX) tumor models are known to maintain the genomic and phenotypic profiles, including the histopathological structures, of the parental tumors. On the other hand, unique enrichment of single-nucleotide variants or copy number aberrations has been reported in several types of tumors. However, an understanding of endometrial carcinoma PDXs is limited. The purpose of the present study was to clarify the presence or absence of the molecular properties of endometrial carcinomas in PDXs passaged up to eight times. Established PDXs of endometrioid carcinomas maintained their histopathological characteristics, but those of carcinosarcomas predominantly consisted of sarcomatous components when compared to the parental tumors. Alterations in the proportion of cells with positive/negative immunohistochemical staining for estrogen receptor, PTEN, PAX8, and PAX2 were observed, whereas the proportions of cells with AE1/AE3, TP53, ARID1A, PMS2, and MSH6 staining were unchanged. Variants of cancer-associated genes were compared between PDXs and parental tumors. Mutations in POLE and a frameshift deletion in BRCA1 were observed in the parental tumor tissue in each of the six cases, and additional genomic alterations, which were not apparently related to histopathological and immunohistochemical alterations, were found in the PDXs of these cases. The genomic and phenotypic alterations observed between endometrial carcinoma PDXs and parental tumors were partly associated with endometrial cancer-specific characteristics related to cellular differentiation and gene mutations.
Collapse
Affiliation(s)
- Toshio Imai
- Central Animal Division, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan.
| | - Hiroshi Yoshida
- Department of Diagnostic Pathology, National Cancer Center Hospital, Tokyo, Japan
| | - Yukino Machida
- Central Animal Division, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Mizuki Kuramochi
- Central Animal Division, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Hitoshi Ichikawa
- Department of Clinical Genomics, National Cancer Center Research Institute, Tokyo, Japan
| | - Takashi Kubo
- Department of Clinical Genomics, National Cancer Center Research Institute, Tokyo, Japan
| | - Mami Takahashi
- Central Animal Division, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Tomoyasu Kato
- Department of Gynecology, National Cancer Center Hospital, Tokyo, Japan
| |
Collapse
|
47
|
Khayatan D, Hussain A, Tebyaniyan H. Exploring animal models in oral cancer research and clinical intervention: A critical review. Vet Med Sci 2023. [PMID: 37196179 DOI: 10.1002/vms3.1161] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 03/27/2023] [Accepted: 04/28/2023] [Indexed: 05/19/2023] Open
Abstract
Cancer is a leading cause of death worldwide, but advances in treatment, early detection, and prevention have helped to reduce its impact. To translate cancer research findings into clinical interventions for patients, appropriate animal experimental models, particularly in oral cancer therapy, can be helpful. In vitro experiments using animal or human cells can provide insight into cancer's biochemical pathways. This review discusses the various animal models used in recent years for research and clinical intervention in oral cancer, along with their advantages and disadvantages. We highlight the advantages and limitations of the used animal models in oral cancer research and therapy by searching the terms of animal models, oral cancer, oral cancer therapy, oral cancer research, and animals to find all relevant publications during 2010-2023. Mouse models, widely used in cancer research, can help us understand protein and gene functions in vivo and molecular pathways more deeply. To induce cancer in rodents, xenografts are often used, but companion animals with spontaneous tumours are underutilized for rapid advancement in human and veterinary cancer treatments. Like humans with cancer, companion animals exhibit biological behaviour, treatment responses, and cytotoxic agent responses similar to humans. In companion animal models, disease progression is more rapid, and the animals have a shorter lifespan. Animal models allow researchers to study how immune cells interact with cancer cells and how they can be targeted specifically. Additionally, animal models have been extensively used in research on oral cancers, so researchers can use existing knowledge and tools to better understand oral cancers using animal models.
Collapse
Affiliation(s)
- Danial Khayatan
- GI Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Ahmed Hussain
- School of Dentistry, Edmonton Clinic Health Academy (ECHA), University of Alberta, Edmonton, Canada
| | - Hamid Tebyaniyan
- Department of Science and Research, Islimic Azade University, Tehran, Iran
| |
Collapse
|
48
|
Foss A, Pathania M. Pediatric Glioma Models Provide Insights into Tumor Development and Future Therapeutic Strategies. Dev Neurosci 2023; 46:22-43. [PMID: 37231843 DOI: 10.1159/000531040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 05/09/2023] [Indexed: 05/27/2023] Open
Abstract
In depth study of pediatric gliomas has been hampered due to difficulties in accessing patient tissue and a lack of clinically representative tumor models. Over the last decade, however, profiling of carefully curated cohorts of pediatric tumors has identified genetic drivers that molecularly segregate pediatric gliomas from adult gliomas. This information has inspired the development of a new set of powerful in vitro and in vivo tumor models that can aid in identifying pediatric-specific oncogenic mechanisms and tumor microenvironment interactions. Single-cell analyses of both human tumors and these newly developed models have revealed that pediatric gliomas arise from spatiotemporally discrete neural progenitor populations in which developmental programs have become dysregulated. Pediatric high-grade gliomas also harbor distinct sets of co-segregating genetic and epigenetic alterations, often accompanied by unique features within the tumor microenvironment. The development of these novel tools and data resources has led to insights into the biology and heterogeneity of these tumors, including identification of distinctive sets of driver mutations, developmentally restricted cells of origin, recognizable patterns of tumor progression, characteristic immune environments, and tumor hijacking of normal microenvironmental and neural programs. As concerted efforts have broadened our understanding of these tumors, new therapeutic vulnerabilities have been identified, and for the first time, promising new strategies are being evaluated in the preclinical and clinical settings. Even so, dedicated and sustained collaborative efforts are necessary to refine our knowledge and bring these new strategies into general clinical use. In this review, we will discuss the range of currently available glioma models, the way in which they have each contributed to recent developments in the field, their benefits and drawbacks for addressing specific research questions, and their future utility in advancing biological understanding and treatment of pediatric glioma.
Collapse
Affiliation(s)
- Amelia Foss
- Department of Oncology and the Milner Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
- CRUK Children's Brain Tumour Centre of Excellence, University of Cambridge, Cambridge, UK
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Manav Pathania
- Department of Oncology and the Milner Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
- CRUK Children's Brain Tumour Centre of Excellence, University of Cambridge, Cambridge, UK
| |
Collapse
|
49
|
Patel P, Nandi A, Verma SK, Kaushik N, Suar M, Choi EH, Kaushik NK. Zebrafish-based platform for emerging bio-contaminants and virus inactivation research. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 872:162197. [PMID: 36781138 PMCID: PMC9922160 DOI: 10.1016/j.scitotenv.2023.162197] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/23/2023] [Accepted: 02/08/2023] [Indexed: 05/27/2023]
Abstract
Emerging bio-contaminants such as viruses have affected health and environment settings of every country. Viruses are the minuscule entities resulting in severe contagious diseases like SARS, MERS, Ebola, and avian influenza. Recent epidemic like the SARS-CoV-2, the virus has undergone mutations strengthen them and allowing to escape from the remedies. Comprehensive knowledge of viruses is essential for the development of targeted therapeutic and vaccination treatments. Animal models mimicking human biology like non-human primates, rats, mice, and rabbits offer competitive advantage to assess risk of viral infections, chemical toxins, nanoparticles, and microbes. However, their economic maintenance has always been an issue. Furthermore, the redundancy of experimental results due to aforementioned aspects is also in examine. Hence, exploration for the alternative animal models is crucial for risk assessments. The current review examines zebrafish traits and explores the possibilities to monitor emerging bio-contaminants. Additionally, a comprehensive picture of the bio contaminant and virus particle invasion and abatement mechanisms in zebrafish and human cells is presented. Moreover, a zebrafish model to investigate the emerging viruses such as coronaviridae and poxviridae has been suggested.
Collapse
Affiliation(s)
- Paritosh Patel
- Plasma Bioscience Research Center, Department of Electrical and Biological Physics, Kwangwoon University, 01897 Seoul, South Korea
| | - Aditya Nandi
- School of Biotechnology, KIIT University, Bhubaneswar 751024, Odisha, India
| | - Suresh K Verma
- School of Biotechnology, KIIT University, Bhubaneswar 751024, Odisha, India; Condensed Matter Theory Group, Materials Theory Division, Department of Physics and Astronomy, Uppsala University, Box 516, SE-751 20 Uppsala, Sweden
| | - Neha Kaushik
- Department of Biotechnology, College of Engineering, The University of Suwon, 18323 Hwaseong, Republic of Korea
| | - Mrutyunjay Suar
- School of Biotechnology, KIIT University, Bhubaneswar 751024, Odisha, India
| | - Eun Ha Choi
- Plasma Bioscience Research Center, Department of Electrical and Biological Physics, Kwangwoon University, 01897 Seoul, South Korea.
| | - Nagendra Kumar Kaushik
- Plasma Bioscience Research Center, Department of Electrical and Biological Physics, Kwangwoon University, 01897 Seoul, South Korea.
| |
Collapse
|
50
|
Ding T, Yu Y, Pan X, Chen H. Establishment of humanized mice and its application progress in cancer immunotherapy. Immunotherapy 2023; 15:679-697. [PMID: 37096919 DOI: 10.2217/imt-2022-0148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2023] Open
Abstract
The current high prevalence of malignant tumors has attracted considerable attention, and treating advanced malignancies is becoming increasingly difficult. Although immunotherapy is a hopeful alternative, it is effective in only a few people. Thus, development of preclinical animal models is needed. Humanized xenotransplantation mouse models can help with selecting treatment protocols, evaluating curative effects and assessing prognosis. This review discusses the establishment of humanized mouse models and their application prospects in cancer immunotherapy to identify tailored therapies for individual patients.
Collapse
Affiliation(s)
- Tianlong Ding
- The Second Clinical Medical College, Lanzhou University, Lanzhou, 730030, PR China
- Department of Tumor Surgery, Lanzhou University Second Hospital, Lanzhou, 730030, PR China
| | - Yang Yu
- The Second Clinical Medical College, Lanzhou University, Lanzhou, 730030, PR China
| | - Xiaoyuan Pan
- Department of Vision Rehabilitation, Gansu Province Hospital Rehabilitation Center, Lanzhou, 730030, PR China
| | - Hao Chen
- Department of Tumor Surgery, Lanzhou University Second Hospital, Lanzhou, 730030, PR China
- Key Laboratory of Digestive System Tumors, Lanzhou University Second Hospital, Lanzhou, 730030, PR China
| |
Collapse
|