1
|
Alegre ML, Atkinson C, Issa F, Valujskikh A, Zhang ZJ. BEST PRACTICES OF HEART TRANSPLANTATION IN MICE. Am J Transplant 2025:S1600-6135(25)00217-5. [PMID: 40252924 DOI: 10.1016/j.ajt.2025.04.012] [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: 01/09/2025] [Revised: 03/21/2025] [Accepted: 04/12/2025] [Indexed: 04/21/2025]
Abstract
Heart transplantation in mice has served as a reliable in vivo model in transplant research worldwide for more than half a century. It is not only useful for addressing cardiac graft-specific questions but also provides mechanistic insights and therapeutic strategies that have broad impact across all solid organ transplants. Compared to other mouse models of solid organ transplantation, such as kidney, lung, or small intestine transplants, the surgical techniques to perform mouse heart transplantation (mHT) are relatively easy to master, and the graft heartbeat offers a simple means to evaluate transplant viability. However, as with other in vivo mouse models, mHT has distinct strengths and limitations. Multiple factors can influence the accuracy and reproducibility of the results, including microsurgical techniques and microsurgeons' skills, post-op monitoring methodologies, mouse strain combinations, sex/age. As innovative biotechnologies continue to emerge, the future holds many opportunities for preclinical research utilizing the mHT model. It is therefore imperative to provide the field with optimized mHT protocols and maintain standard reporting requirements. This minireview provides a concise summary and recommendations for standardized practices to ensure the accuracy, reproducibility, and translational value of findings generated from mHT model.
Collapse
Affiliation(s)
- Maria-Luisa Alegre
- Department of Medicine, Section of Rheumatology, The University of Chicago, Chicago, IL, USA
| | - Carl Atkinson
- Comprehensive Transplant Center, Northwestern University Feinberg School of Medicine; Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Fadi Issa
- Nuffield Department of Surgical Sciences, University of Oxford, United Kingdom
| | - Anna Valujskikh
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland, OH; Cleveland Clinic Lerner College of Medicine, Cleveland, OH, United States
| | - Zheng Jenny Zhang
- Comprehensive Transplant Center, Northwestern University Feinberg School of Medicine; Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| |
Collapse
|
2
|
Craft K, Amanor A, Barnett I, Donaldson C, Anegon I, Madduri S, Tang Q, Bility MT. Can Humanized Immune System Mouse and Rat Models Accelerate the Development of Cytomegalovirus-Based Vaccines Against Infectious Diseases and Cancers? Int J Mol Sci 2025; 26:3082. [PMID: 40243710 PMCID: PMC11988357 DOI: 10.3390/ijms26073082] [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: 03/05/2025] [Revised: 03/20/2025] [Accepted: 03/22/2025] [Indexed: 04/18/2025] Open
Abstract
Over the past three decades, immunodeficient mouse models carrying human immune cells, with or without human lymphoid tissues, termed humanized immune system (HIS) rodent models, have been developed to recapitulate the human immune system and associated immune responses. HIS mouse models have successfully modeled many human-restricted viral infections, including those caused by human cytomegalovirus (HCMV) and human immunodeficiency virus (HIV). HIS mouse models have also been used to model human cancer immunobiology, which exhibits differences from murine cancers in traditional mouse models. Variants of HIS mouse models that carry human liver cells, lung tissue, skin tissue, or human patient-derived tumor xenografts and human hematopoietic stem cells-derived-human immune cells with or without lymphoid tissue xenografts have been developed to probe human immune responses to infections and human tumors. HCMV-based vaccines are human-restricted, which poses limitations for mechanistic and efficacy studies using traditional animal models. The HCMV-based vaccine approach is a promising vaccine strategy as it induces robust effector memory T cell responses that may be critical in preventing and rapidly controlling persistent viral infections and cancers. Here, we review novel HIS mouse models with robust human immune cell development and primary and secondary lymphoid tissues that could address many of the limitations of HIS mice in their use as animal models for HCMV-based vaccine research. We also reviewed novel HIS rat models, which could allow long-term (greater than one year) vaccinology studies and better recapitulate human pathophysiology. Translating laboratory research findings to clinical application is a significant bottleneck in vaccine development; HIS rodents and related variants that more accurately model human immunology and diseases could increase the translatability of research findings.
Collapse
Affiliation(s)
- Kaci Craft
- Department of Microbiology, Howard University College of Medicine, Washington, DC 20059, USA; (K.C.); (A.A.); (I.B.); (C.D.); (Q.T.)
| | - Athina Amanor
- Department of Microbiology, Howard University College of Medicine, Washington, DC 20059, USA; (K.C.); (A.A.); (I.B.); (C.D.); (Q.T.)
| | - Ian Barnett
- Department of Microbiology, Howard University College of Medicine, Washington, DC 20059, USA; (K.C.); (A.A.); (I.B.); (C.D.); (Q.T.)
| | - Clarke Donaldson
- Department of Microbiology, Howard University College of Medicine, Washington, DC 20059, USA; (K.C.); (A.A.); (I.B.); (C.D.); (Q.T.)
| | - Ignacio Anegon
- Nantes Université, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, F-44000 Nantes, France;
| | - Srinivas Madduri
- Bioengineering and Neuroregeneration Laboratory, Department of Surgery, University of Geneva, 1211 Geneva, Switzerland;
| | - Qiyi Tang
- Department of Microbiology, Howard University College of Medicine, Washington, DC 20059, USA; (K.C.); (A.A.); (I.B.); (C.D.); (Q.T.)
| | - Moses T. Bility
- Department of Microbiology, Howard University College of Medicine, Washington, DC 20059, USA; (K.C.); (A.A.); (I.B.); (C.D.); (Q.T.)
| |
Collapse
|
3
|
Gantier M, Ménoret S, Fourrier A, Delbos F, Nguyen TH, Anegon I. Human pluripotent stem cell-derived hepatic progenitors exhibit a partially hypoimmunogenic phenotype and actively inhibit immune responses. Front Immunol 2025; 16:1507317. [PMID: 40070824 PMCID: PMC11893836 DOI: 10.3389/fimmu.2025.1507317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 02/03/2025] [Indexed: 03/14/2025] Open
Abstract
Introduction GStemHep cells are human cryopreserved hepatic progenitors derived from pluripotent of stem cells (GStem cells) using a cGMP-compliant protocol. They were highly effective in rescuing mice from acute liver failure. Methods The objective of this study was to analyze the immunogenicity and immunoregulatory properties of GStemHep cells. Results As compared to GStem cells, GStemHep cells showed complete loss of HLA-I (ABC) and they lacked of expression of HLA-II, HLA-G, HLA-E and PD-L1. GStemHep cells also showed increased expression of CD47, maintained high expression of indoleamine 2,3-dioxygenase (IDO) and heme oxygenase-1 (HO-1) and reduced expression of CD200. In comparison with GStem cells, GStemHep cultured in inflammatory conditions increased the expression of PD-L1, CD200, HO-1, HLA-E, CD47 and HLA-I (ABC) as well as maintained expression of IDO and were negative for HLA-II and HLA-G. GStemHep culture in basal or inflammatory conditions has a low or absent immunogenic activity on T cells, associated to a suppressive effect on proliferation partially mediated by IDO. We observed phagocytosis of GStemHep by macrophages that was partially inhibited by CD47 expression. NK cells were activated by resting GStemHep cells. Upon culture in inflammatory conditions that induced expression of HLA-I molecules in GStemHep cells NK cell activation was reduced. Thus, GStemHep cells are partially hypoimmune cells due to the expression of several immune checkpoint inhibitors and the absence of HLA-I molecules. In inflammatory conditions, the expression of several of these molecules was increased but also of HLA-I that could be immunogenic for T cells but it was inhibitory for NK cells. Discussion GStemHep cells show a favorable immunological profile for their use as allogeneic off-the shelf treatment of liver diseases with loss of hepatocyte function.
Collapse
Affiliation(s)
| | - Séverine Ménoret
- Nantes Université, Inserm, CNRS, SFR Santé, Inserm UMS 016 CNRS UMS 3556, Nantes, France
- Nantes Université, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
| | | | | | | | - Ignacio Anegon
- Nantes Université, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
| |
Collapse
|
4
|
Nakano K, Goto M, Fukuda S, Yanobu-Takanashi R, Yabe SG, Shimizu Y, Sakuma T, Yamamoto T, Shimoda M, Okochi H, Takahashi R, Okamura T. A Novel Immunodeficient Hyperglycemic Mouse Carrying the Ins1 Akita Mutation for Xenogeneic Islet Cell Transplantation. Transplantation 2025; 109:e81-e91. [PMID: 39104009 PMCID: PMC11745600 DOI: 10.1097/tp.0000000000005152] [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: 02/14/2024] [Revised: 06/04/2024] [Accepted: 06/12/2024] [Indexed: 08/07/2024]
Abstract
BACKGROUND For patients who have difficulty controlling blood glucose even with insulin administration, xenogeneic islet cells, including human stem cell-derived pancreatic islets (hSC-islet) and porcine islets, have garnered attention as potential solutions to challenges associated with donor shortages. For the development of diabetes treatment modalities that use cell transplantation therapy, it is essential to evaluate the efficacy and safety of transplanted cells using experimental animals over the long term. METHODS We developed permanent diabetic immune-deficient mice by introducing the Akita (C96Y) mutation into the rodent-specific Insulin1 gene of NOD/Shi-scid IL2rγc null (NOG) mice ( Ins1 C96Y/C96Y NOG). Their body weight, nonfasting blood glucose, and survival were measured from 4 wk of age. Insulin sensitivity was assessed via tolerance tests. To elucidate the utility of these mice in xenotransplantation experiments, we transplanted hSC-islet cells or porcine islets under the kidney capsules of these mice. RESULTS All male and female homozygous mice exhibited persistent severe hyperglycemia associated with β-cell depletion as early as 4 wk of age and exhibited normal insulin sensitivity. These mice could be stably engrafted with hSC-islets, and the mice that received porcine islet grafts promptly exhibited lowered blood glucose levels, maintaining blood glucose levels below the normal glucose range for at least 52 wk posttransplantation. CONCLUSIONS The Ins1C96Y/C96Y NOG mouse model provides an effective platform to assess both the efficacy and safety of long-term xenograft engraftment without the interference of their immune responses. This study is expected to contribute essential basic information for the clinical application of islet cell transplantation.
Collapse
Affiliation(s)
- Kenta Nakano
- Department of Laboratory Animal Medicine, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Motohito Goto
- Animal Resource Technical Research Center, Central Institute for Experimental Medicine and Life Science, Kawasaki, Japan
| | - Satsuki Fukuda
- Department of Regenerative Medicine, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Rieko Yanobu-Takanashi
- Department of Laboratory Animal Medicine, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Shigeharu G. Yabe
- Department of Regenerative Medicine, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Yukiko Shimizu
- Department of Laboratory Animal Medicine, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
- Department of Pediatrics, Juntendo University School of Medicine, Tokyo, Japan
| | - Tetsushi Sakuma
- Division of Integrated Sciences for Life, Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, Japan
| | - Takashi Yamamoto
- Division of Integrated Sciences for Life, Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, Japan
| | - Masayuki Shimoda
- Department of Pancreatic Islet Cell Transplantation, Diabetes Research Center, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Hitoshi Okochi
- Department of Regenerative Medicine, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Riichi Takahashi
- Animal Resource Technical Research Center, Central Institute for Experimental Medicine and Life Science, Kawasaki, Japan
| | - Tadashi Okamura
- Department of Laboratory Animal Medicine, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
- Laboratory of Laboratory Animal Science and Medicine, School of Veterinary Medicine, Kitasato University, Tokyo, Japan
| |
Collapse
|
5
|
Rodríguez E, Muñoz-Fontela C, Escudero-Pérez B. Filovirus Infection in Humanized Mouse Models. Methods Mol Biol 2025; 2877:213-226. [PMID: 39585624 DOI: 10.1007/978-1-0716-4256-6_15] [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: 11/26/2024]
Abstract
Since the discovery of filoviruses in 1967, there have been more than 40 outbreaks with high case-fatality rates causing more than 30,000 deaths in humans. Filovirus disease (FVD) involves the dysregulation of many host physiological processes. While many advances in the field have taken place since the first outbreaks, the dearth of small animal models that translate the features observed during FVD in humans has limited our understanding of the pathology. Here, we describe the generation and use of human immune system (HIS) mice as a preclinical model to investigate FVD in a human-like immune environment.
Collapse
Affiliation(s)
- Estefanía Rodríguez
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- German Center for Infection Research (DZIF), Partner Site Hamburg-Luebeck-Borstel-Reims, Hamburg, Germany
| | - César Muñoz-Fontela
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- German Center for Infection Research (DZIF), Partner Site Hamburg-Luebeck-Borstel-Reims, Hamburg, Germany
| | - Beatriz Escudero-Pérez
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany.
- German Center for Infection Research (DZIF), Partner Site Hamburg-Luebeck-Borstel-Reims, Hamburg, Germany.
| |
Collapse
|
6
|
Yamaguchi N, Horio E, Sonoda J, Yamagishi M, Miyakawa S, Murakami F, Hasegawa H, Katahira Y, Mizoguchi I, Fujii Y, Chikazu D, Yoshimoto T. Immortalization of Mesenchymal Stem Cells for Application in Regenerative Medicine and Their Potential Risks of Tumorigenesis. Int J Mol Sci 2024; 25:13562. [PMID: 39769322 PMCID: PMC11676347 DOI: 10.3390/ijms252413562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 12/13/2024] [Accepted: 12/16/2024] [Indexed: 01/11/2025] Open
Abstract
Regenerative medicine utilizes stem cells to repair damaged tissues by replacing them with their differentiated cells and activating the body's inherent regenerative abilities. Mesenchymal stem cells (MSCs) are adult stem cells that possess tissue repair and regenerative capabilities and immunomodulatory properties with a much lower risk of tumorigenicity, making them a focus of numerous clinical trials worldwide. MSCs primarily exert their therapeutic effects through paracrine effects via secreted factors, such as cytokines and exosomes. This has led to increasing interest in cell-free therapy, where only the conditioned medium (also called secretome) from MSC cultures is used for regenerative applications. However, MSCs face certain limitations, including cellular senescence, scarcity, donor heterogeneity, complexity, short survival post-implantation, and regulatory and ethics hurdles. To address these challenges, various types of immortalized MSCs (ImMSCs) capable of indefinite expansion have been developed. These cells offer significant promise and essential tools as a reliable source for both cell-based and cell-free therapies with the aim of translating them into practical medicine. However, the process of immortalization, often involving the transduction of immortalizing genes, poses potential risks of genetic instability and resultant malignant transformation. Cell-free therapy is particularly attractive, as it circumvents the risks of tumorigenicity and ethical concerns associated with live cell therapies. Rigorous safety tests, such as monitoring chromosomal abnormalities, are critical to ensure safety. Technologies like inducible or suicide genes may allow for the controlled proliferation of MSCs and induce apoptosis after their therapeutic task is completed. This review highlights recent advancements in the immortalization of MSCs and the associated risks of tumorigenesis.
Collapse
Affiliation(s)
- Natsuki Yamaguchi
- Department of Immunoregulation, Institute of Medical Science, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku-ku, Tokyo 160-8402, Japan
| | - Eri Horio
- Department of Immunoregulation, Institute of Medical Science, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku-ku, Tokyo 160-8402, Japan
| | - Jukito Sonoda
- Department of Immunoregulation, Institute of Medical Science, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku-ku, Tokyo 160-8402, Japan
| | - Miu Yamagishi
- Department of Immunoregulation, Institute of Medical Science, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku-ku, Tokyo 160-8402, Japan
| | - Satomi Miyakawa
- Department of Immunoregulation, Institute of Medical Science, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku-ku, Tokyo 160-8402, Japan
| | - Fumihiro Murakami
- Department of Immunoregulation, Institute of Medical Science, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku-ku, Tokyo 160-8402, Japan
| | - Hideaki Hasegawa
- Department of Immunoregulation, Institute of Medical Science, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku-ku, Tokyo 160-8402, Japan
| | - Yasuhiro Katahira
- Department of Immunoregulation, Institute of Medical Science, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku-ku, Tokyo 160-8402, Japan
| | - Izuru Mizoguchi
- Department of Immunoregulation, Institute of Medical Science, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku-ku, Tokyo 160-8402, Japan
| | - Yasuyuki Fujii
- Department of Oral and Maxillofacial Surgery, Tokyo Medical University, 6-7-1 Nishishinjuku, Shinjuku-ku, Tokyo 160-0023, Japan
| | - Daichi Chikazu
- Department of Oral and Maxillofacial Surgery, Tokyo Medical University, 6-7-1 Nishishinjuku, Shinjuku-ku, Tokyo 160-0023, Japan
| | - Takayuki Yoshimoto
- Department of Immunoregulation, Institute of Medical Science, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku-ku, Tokyo 160-8402, Japan
| |
Collapse
|
7
|
Yu S, Xie J, Li PH, Chen Y, Tang IY, Lin X. Therapeutic potential of interleukin-17 neutralization in a novel humanized mouse model of Sjögren's disease. Pharmacol Res 2024; 210:107524. [PMID: 39617280 DOI: 10.1016/j.phrs.2024.107524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 11/26/2024] [Accepted: 11/26/2024] [Indexed: 12/06/2024]
Abstract
Sjögren's disease (SjD) is a chronic autoimmune disease, in which the immune system targets exocrine glands and leads to dryness symptoms. There is an increasing need to develop novel therapeutic approach as the treatment plan has not been changed in the past decade. However, findings in mouse model may not be directly applied in patients, given the substantial differences of immune system between human and mice. In the present study, using antigens derived from human salivary A-253 cells, we established experimental Sjögren's syndrome (ESS) in mice with human immune system (HIS). HIS-ESS mice exhibited key features of human disease, including salivary hypofunction, increased serum levels of autoantibodies and tissue destruction in the salivary glands. Phenotypic analysis revealed enhanced effector B and T cell subsets, including Th1, Th17 and T follicular helper (Tfh) cells in HIS-ESS mice, while multiplex imaging analysis suggested enlarged B cell follicles and expanded memory B cells. IL-17 neutralization therapy significantly ameliorated disease pathology at both acute and chronic stages, in which B cells were mainly affected, to the less extent Th1 and Tfh cells in HIS-ESS mice. Together, HIS-ESS mouse model highly recapitulated SjD features and immunopathogenesis, which may serve as a useful tool in drug screening and pre-clinical studies.
Collapse
Affiliation(s)
- Sulan Yu
- School of Chinese Medicine, the University of Hong Kong, Hong Kong
| | - Jing Xie
- School of Chinese Medicine, the University of Hong Kong, Hong Kong
| | - Philip Hei Li
- Division of Rheumatology and Clinical Immunology, Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Hong Kong
| | - Yacun Chen
- School of Chinese Medicine, the University of Hong Kong, Hong Kong
| | - Iris Yanki Tang
- School of Chinese Medicine, the University of Hong Kong, Hong Kong
| | - Xiang Lin
- School of Chinese Medicine, the University of Hong Kong, Hong Kong; State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong.
| |
Collapse
|
8
|
Rohban R, Martins CP, Esni F. Advanced therapy to cure diabetes: mission impossible is now possible? Front Cell Dev Biol 2024; 12:1484859. [PMID: 39629270 PMCID: PMC11611888 DOI: 10.3389/fcell.2024.1484859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 11/04/2024] [Indexed: 12/07/2024] Open
Abstract
Cell and Gene therapy are referred to as advanced therapies that represent overlapping fields of regenerative medicine. They have similar therapeutic goals such as to modify cellular identity, improve cell function, or fight a disease. These two therapeutic avenues, however, possess major differences. While cell therapy involves introduction of new cells, gene therapy entails introduction or modification of genes. Furthermore, the aim of cell therapy is often to replace, or repair damaged tissue, whereas gene therapy is used typically as a preventive approach. Diabetes mellitus severely affects the quality of life of afflicted individuals and has various side effects including cardiovascular, ophthalmic disorders, and neuropathy while putting enormous economic pressure on both the healthcare system and the patient. In recent years, great effort has been made to develop cutting-edge therapeutic interventions for diabetes treatment, among which cell and gene therapies stand out. This review aims to highlight various cell- and gene-based therapeutic approaches leading to the generation of new insulin-producing cells as a topmost "panacea" for treating diabetes, while deliberately avoiding a detailed molecular description of these approaches. By doing so, we aim to target readers who are new to the field and wish to get a broad helicopter overview of the historical and current trends of cell- and gene-based approaches in β-cell regeneration.
Collapse
Affiliation(s)
- Rokhsareh Rohban
- Department of Internal Medicine, Division of Hematology, Medical University of Graz, Graz, Austria
| | - Christina P. Martins
- Department of Surgery, Division of Pediatric General and Thoracic Surgery, Children’s Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Farzad Esni
- Department of Surgery, Division of Pediatric General and Thoracic Surgery, Children’s Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
- Department of Developmental Biology, University of Pittsburgh, Pittsburgh, PA, United States
- UPMC Hillman Cancer Center, Pittsburgh, PA, United States
- McGowan Institute for regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
9
|
Clark BJ, Lelos MJ, Loring JF. Advancing Parkinson's disease treatment: cell replacement therapy with neurons derived from pluripotent stem cells. Stem Cells 2024; 42:781-790. [PMID: 38902932 DOI: 10.1093/stmcls/sxae042] [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: 03/11/2024] [Accepted: 05/24/2024] [Indexed: 06/22/2024]
Abstract
The motor symptoms of Parkinson's disease (PD) are caused by the progressive loss of dopamine neurons from the substantia nigra. There are currently no treatments that can slow or reverse the neurodegeneration. To restore the lost neurons, international groups have initiated clinical trials using human embryonic or induced pluripotent stem cells (PSCs) to derive dopamine neuron precursors that are used as transplants to replace the lost neurons. Proof-of-principle experiments in the 1980s and 1990s showed that grafts of fetal ventral mesencephalon, which contains the precursors of the substantial nigra, could, under rare circumstances, reverse symptoms of the disease. Improvements in PSC technology and genomics have inspired researchers to design clinical trials using PSC-derived dopamine neuron precursors as cell replacement therapy for PD. We focus here on 4 such first-in-human clinical trials that have begun in the US, Europe, and Japan. We provide an overview of the sources of PSCs and the methods used to generate cells for transplantation. We discuss pros and cons of strategies for allogeneic, immune-matched, and autologous approaches and novel methods for overcoming rejection by the immune system. We consider challenges for safety and efficacy of the cells for durable engraftment, focusing on the genomics-based quality control methods to assure that the cells will not become cancerous. Finally, since clinical trials like these have never been undertaken before, we comment on the value of cooperation among rivals to contribute to advancements that will finally provide relief for the millions suffering from the symptoms of PD.
Collapse
Affiliation(s)
- Branden J Clark
- Department of Biomedical Engineering, UC Irvine, Irvine, CA 92697, United States
| | - Mariah J Lelos
- School of Biosciences, Museum Avenue, Cardiff University, Cardiff, CF10 3AX, United Kingdom
| | - Jeanne F Loring
- Department of Molecular Medicine, Scripps Research, La Jolla, CA 92030, United States
| |
Collapse
|
10
|
Ménoret S, Renart-Depontieu F, Martin G, Thiam K, Anegon I. Efficient generation of human immune system rats using human CD34 + cells. Stem Cell Reports 2024; 19:1255-1263. [PMID: 39151431 PMCID: PMC11411320 DOI: 10.1016/j.stemcr.2024.07.005] [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: 10/02/2023] [Revised: 07/20/2024] [Accepted: 07/21/2024] [Indexed: 08/19/2024] Open
Abstract
Human immune system (HIS) mice generated using human CD34+ hematopoietic stem cells serve as a pivotal model for the in vivo evaluation of immunotherapies for humans. Yet, HIS mice possess certain limitations. Rats, due to their size and comprehensive immune system, hold promise for translational experiments. Here, we describe an efficacious method for long-term immune humanization, through intrahepatic injection of hCD34+ cells in newborn immunodeficient rats expressing human SIRPα. In contrast to HIS mice and similar to humans, HIS rats showed in blood a predominance of T cells, followed by B cells. Immune humanization was also high in central and secondary lymphoid organs. HIS rats treated with the anti-human CD3 antibody were depleted of human T cells, and human cytokines were detected in sera. We describe for the first time a method to efficiently generate HIS rats. HIS rats have the potential to be a useful model for translational immunology.
Collapse
Affiliation(s)
- Séverine Ménoret
- Nantes Université, CHU Nantes, Inserm, CNRS, SFR Santé, Inserm UMS 016, CNRS UMS 3556, F-44000 Nantes, France; INSERM, Centre de Recherche en Transplantation et Immunologie UMR1064, Nantes Université, Nantes, France.
| | | | | | | | - Ignacio Anegon
- INSERM, Centre de Recherche en Transplantation et Immunologie UMR1064, Nantes Université, Nantes, France.
| |
Collapse
|
11
|
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
|
12
|
Kitano M, Ohnishi H, Makino A, Miyamoto T, Hayashi Y, Mizuno K, Kaba S, Kawai Y, Kojima T, Kishimoto Y, Yamamoto N, Tomonaga K, Omori K. An Infection Model for SARS-CoV-2 Using Rat Transplanted with hiPSC-Airway Epithelial Cells. Tissue Eng Part A 2024. [PMID: 38832872 DOI: 10.1089/ten.tea.2024.0016] [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: 06/06/2024] Open
Abstract
Investigating the infection mechanism of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in the airway epithelium and developing effective defense strategies against infection are important. To achieve this, establishing appropriate infection models is crucial. Therefore, various in vitro models, such as cell lines and primary cultures, and in vivo models involving animals that exhibit SARS-CoV-2 infection and genetically humanized animals have been used as animal models. However, no animal model has been established that allows infection experiments with human cells under the physiological environment of airway epithelia. Therefore, we aimed to establish a novel animal model that enables infection experiments using human cells. Human induced pluripotent stem cell-derived airway epithelial cell-transplanted nude rats (hiPSC-AEC rats) were used, and infection studies were performed by spraying lentiviral pseudoviruses containing SARS-CoV-2 spike protein and the GFP gene on the tracheae. After infection, immunohistochemical analyses revealed the existence of GFP-positive-infected transplanted cells in the epithelial and submucosal layers. In this study, a SARS-CoV-2 infection animal model including human cells was established mimicking infection through respiration, and we demonstrated that the hiPSC-AEC rat could be used as an animal model for basic research and the development of therapeutic methods for human-specific respiratory infectious diseases.
Collapse
Affiliation(s)
- Masayuki Kitano
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of medicine, Kyoto University, Kyoto City, Japan
| | - Hiroe Ohnishi
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of medicine, Kyoto University, Kyoto City, Japan
| | - Akiko Makino
- Department of Virus Research, Institute for Life and Medical Sciences, Kyoto University, Kyoto City, Japan
| | - Tatsuo Miyamoto
- Department of Molecular and Cellular Physiology, Research Institute for Cell Design Medical Science, Graduate School of Medicine, Yamaguchi University, Ube City, Japan
| | - Yasuyuki Hayashi
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of medicine, Kyoto University, Kyoto City, Japan
| | - Keisuke Mizuno
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of medicine, Kyoto University, Kyoto City, Japan
| | - Shinji Kaba
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of medicine, Kyoto University, Kyoto City, Japan
| | - Yoshitaka Kawai
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of medicine, Kyoto University, Kyoto City, Japan
| | - Tsuyoshi Kojima
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of medicine, Kyoto University, Kyoto City, Japan
| | - Yo Kishimoto
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of medicine, Kyoto University, Kyoto City, Japan
| | - Norio Yamamoto
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of medicine, Kyoto University, Kyoto City, Japan
- Department of Otolaryngology, Kobe City Medical Center General Hospital, Kobe city, Japan
| | - Keizo Tomonaga
- Department of Virus Research, Institute for Life and Medical Sciences, Kyoto University, Kyoto City, Japan
| | - Koichi Omori
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of medicine, Kyoto University, Kyoto City, Japan
| |
Collapse
|
13
|
Bernal-Conde LD, Peña-Martínez V, Morato-Torres CA, Ramos-Acevedo R, Arias-Carrión Ó, Padilla-Godínez FJ, Delgado-González A, Palomero-Rivero M, Collazo-Navarrete O, Soto-Rojas LO, Gómez-Chavarín M, Schüle B, Guerra-Crespo M. Alpha-Synuclein Gene Alterations Modulate Tyrosine Hydroxylase in Human iPSC-Derived Neurons in a Parkinson's Disease Animal Model. Life (Basel) 2024; 14:728. [PMID: 38929711 PMCID: PMC11204703 DOI: 10.3390/life14060728] [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: 05/05/2024] [Revised: 05/27/2024] [Accepted: 06/03/2024] [Indexed: 06/28/2024] Open
Abstract
Parkinson's disease (PD) caused by SNCA gene triplication (3XSNCA) leads to early onset, rapid progression, and often dementia. Understanding the impact of 3XSNCA and its absence is crucial. This study investigates the differentiation of human induced pluripotent stem cell (hiPSC)-derived floor-plate progenitors into dopaminergic neurons. Three different genotypes were evaluated in this study: patient-derived hiPSCs with 3XSNCA, a gene-edited isogenic line with a frame-shift mutation on all SNCA alleles (SNCA 4KO), and a normal wild-type control. Our aim was to assess how the substantia nigra pars compacta (SNpc) microenvironment, damaged by 6-hydroxydopamine (6-OHDA), influences tyrosine hydroxylase-positive (Th+) neuron differentiation in these genetic variations. This study confirms successful in vitro differentiation into neuronal lineage in all cell lines. However, the SNCA 4KO line showed unusual LIM homeobox transcription factor 1 alpha (Lmx1a) extranuclear distribution. Crucially, both 3XSNCA and SNCA 4KO lines had reduced Th+ neuron expression, despite initial successful neuronal differentiation after two months post-transplantation. This indicates that while the SNpc environment supports early neuronal survival, SNCA gene alterations-either amplification or knock-out-negatively impact Th+ dopaminergic neuron maturation. These findings highlight SNCA's critical role in PD and underscore the value of hiPSC models in studying neurodegenerative diseases.
Collapse
Affiliation(s)
- Luis Daniel Bernal-Conde
- Laboratory of Regenerative Medicine, Physiology Department, Faculty of Medicine, National Autonomous University of Mexico, Mexico City 04510, Mexico; (L.D.B.-C.); (V.P.-M.); (C.A.M.-T.); (R.R.-A.); (F.J.P.-G.); (A.D.-G.); (M.G.-C.)
- Molecular Neuropathology Department, Neuroscience Division, Institute of Cell Physiology, National Autonomous University of Mexico, Mexico City 04510, Mexico
| | - Verónica Peña-Martínez
- Laboratory of Regenerative Medicine, Physiology Department, Faculty of Medicine, National Autonomous University of Mexico, Mexico City 04510, Mexico; (L.D.B.-C.); (V.P.-M.); (C.A.M.-T.); (R.R.-A.); (F.J.P.-G.); (A.D.-G.); (M.G.-C.)
- Molecular Neuropathology Department, Neuroscience Division, Institute of Cell Physiology, National Autonomous University of Mexico, Mexico City 04510, Mexico
| | - C. Alejandra Morato-Torres
- Laboratory of Regenerative Medicine, Physiology Department, Faculty of Medicine, National Autonomous University of Mexico, Mexico City 04510, Mexico; (L.D.B.-C.); (V.P.-M.); (C.A.M.-T.); (R.R.-A.); (F.J.P.-G.); (A.D.-G.); (M.G.-C.)
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94304, USA;
| | - Rodrigo Ramos-Acevedo
- Laboratory of Regenerative Medicine, Physiology Department, Faculty of Medicine, National Autonomous University of Mexico, Mexico City 04510, Mexico; (L.D.B.-C.); (V.P.-M.); (C.A.M.-T.); (R.R.-A.); (F.J.P.-G.); (A.D.-G.); (M.G.-C.)
- Molecular Neuropathology Department, Neuroscience Division, Institute of Cell Physiology, National Autonomous University of Mexico, Mexico City 04510, Mexico
| | - Óscar Arias-Carrión
- Movement and Sleep Disorders Unit, Dr. Manuel Gea González General Hospital, Mexico City 14080, Mexico;
| | - Francisco J. Padilla-Godínez
- Laboratory of Regenerative Medicine, Physiology Department, Faculty of Medicine, National Autonomous University of Mexico, Mexico City 04510, Mexico; (L.D.B.-C.); (V.P.-M.); (C.A.M.-T.); (R.R.-A.); (F.J.P.-G.); (A.D.-G.); (M.G.-C.)
- Molecular Neuropathology Department, Neuroscience Division, Institute of Cell Physiology, National Autonomous University of Mexico, Mexico City 04510, Mexico
| | - Alexa Delgado-González
- Laboratory of Regenerative Medicine, Physiology Department, Faculty of Medicine, National Autonomous University of Mexico, Mexico City 04510, Mexico; (L.D.B.-C.); (V.P.-M.); (C.A.M.-T.); (R.R.-A.); (F.J.P.-G.); (A.D.-G.); (M.G.-C.)
- Molecular Neuropathology Department, Neuroscience Division, Institute of Cell Physiology, National Autonomous University of Mexico, Mexico City 04510, Mexico
| | - Marcela Palomero-Rivero
- Neurodevelopment and Physiology Department, Neuroscience Division, Institute of Cell Physiology, National Autonomous University of Mexico, Mexico City 04510, Mexico;
| | - Omar Collazo-Navarrete
- National Laboratory of Genomic Resources, Institute of Biomedical Research, National Autonomous University of Mexico, Mexico City 04510, Mexico;
| | - Luis O. Soto-Rojas
- Laboratory of Molecular Pathogenesis, Laboratory 4, Building A4, Medical Surgeon Career, Faculty of Higher Studies Iztacala, National Autonomous University of Mexico, Mexico City 54090, Mexico;
| | - Margarita Gómez-Chavarín
- Laboratory of Regenerative Medicine, Physiology Department, Faculty of Medicine, National Autonomous University of Mexico, Mexico City 04510, Mexico; (L.D.B.-C.); (V.P.-M.); (C.A.M.-T.); (R.R.-A.); (F.J.P.-G.); (A.D.-G.); (M.G.-C.)
| | - Birgitt Schüle
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94304, USA;
| | - Magdalena Guerra-Crespo
- Laboratory of Regenerative Medicine, Physiology Department, Faculty of Medicine, National Autonomous University of Mexico, Mexico City 04510, Mexico; (L.D.B.-C.); (V.P.-M.); (C.A.M.-T.); (R.R.-A.); (F.J.P.-G.); (A.D.-G.); (M.G.-C.)
- Molecular Neuropathology Department, Neuroscience Division, Institute of Cell Physiology, National Autonomous University of Mexico, Mexico City 04510, Mexico
| |
Collapse
|
14
|
Stribbling SM, Beach C, Ryan AJ. Orthotopic and metastatic tumour models in preclinical cancer research. Pharmacol Ther 2024; 257:108631. [PMID: 38467308 PMCID: PMC11781865 DOI: 10.1016/j.pharmthera.2024.108631] [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: 08/17/2023] [Revised: 02/27/2024] [Accepted: 03/08/2024] [Indexed: 03/13/2024]
Abstract
Mouse models of disease play a pivotal role at all stages of cancer drug development. Cell-line derived subcutaneous tumour models are predominant in early drug discovery, but there is growing recognition of the importance of the more complex orthotopic and metastatic tumour models for understanding both target biology in the correct tissue context, and the impact of the tumour microenvironment and the immune system in responses to treatment. The aim of this review is to highlight the value that orthotopic and metastatic models bring to the study of tumour biology and drug development while pointing out those models that are most likely to be encountered in the literature. Important developments in orthotopic models, such as the increasing use of early passage patient material (PDXs, organoids) and humanised mouse models are discussed, as these approaches have the potential to increase the predictive value of preclinical studies, and ultimately improve the success rate of anticancer drugs in clinical trials.
Collapse
Affiliation(s)
- Stephen M Stribbling
- Department of Chemistry, University College London, Gower Street, London WC1E 6BT, UK.
| | - Callum Beach
- Department of Oncology, University of Oxford, ORCRB, Roosevelt Drive, Oxford OX3 7DQ, UK
| | - Anderson J Ryan
- Department of Oncology, University of Oxford, ORCRB, Roosevelt Drive, Oxford OX3 7DQ, UK; Fast Biopharma, Aston Rowant, Oxfordshire, OX49 5SW, UK.
| |
Collapse
|
15
|
Huang Z, Zhang R, Teng Y, Guo J, Zhang H, Wang L, Tang LV, Shi W, Wu Q, Xia L. Nuclear Matrix-associated Protein SMAR1 Attenuated Acute Graft-versus-host Disease by Targeting JAK-STAT Signaling in CD4 + T Cells. Transplantation 2024; 108:e23-e35. [PMID: 37817309 DOI: 10.1097/tp.0000000000004818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/12/2023]
Abstract
BACKGROUND Acute graft-versus-host disease (aGVHD) mediated by alloreactive T cells remains a serious and life-threatening complication of allogeneic hematopoietic cell transplantation (allo-HCT). The contribution of the different CD4 + T helper cell subtypes to the pathogenesis and regulation of aGVHD is a central point in current research. The specialized effector subsets of T cells that differentiate from naive T cells into mature cells are closely related to scaffold/matrix-associated region-1-binding protein (SMAR1). However, the role of SMAR1 in aGVHD is unclear. METHODS Peripheral blood was collected from the patients with or without aGVHD after allo-HCT. The differences in CD4 + T cells transduced with the SMAR1 lentivirus vector and empty vector were analyzed. A humanized aGVHD mouse model was constructed to evaluate the function of SMAR1 in aGVHD. RESULTS The expression of SMAR1 was significantly reduced in the CD4 + T cells from aGVHD patients and related to the occurrence of aGVHD. SMAR1 overexpression in human CD4 + T cells regulated CD4 + T-cell subsets differentiation and inflammatory cytokines secretion and inhibited the Janus kinase/signal transducer and activator of transcription pathway. Moreover, SMAR1 changed chromatin accessibility landscapes and affected the binding motifs of key transcription factors regulating T cells. Additionally, upregulation of SMAR1 expression in CD4 + T cells improved the survival and pathology in a humanized aGVHD mouse model. CONCLUSIONS Our results showed that upregulation of SMAR1 regulated the CD4 + T-cell subpopulation and cytokines secretion and improved survival in a humanized aGVHD mouse model by alleviating inflammation. This study provides a promising therapeutic target for aGVHD.
Collapse
Affiliation(s)
- Zhenli Huang
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ran Zhang
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yao Teng
- Department of Rheumatology and Immunology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jingjing Guo
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hongyong Zhang
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Li Wang
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Liang V Tang
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Shi
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qiuling Wu
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Linghui Xia
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
16
|
Chehelgerdi M, Chehelgerdi M, Khorramian-Ghahfarokhi M, Shafieizadeh M, Mahmoudi E, Eskandari F, Rashidi M, Arshi A, Mokhtari-Farsani A. Comprehensive review of CRISPR-based gene editing: mechanisms, challenges, and applications in cancer therapy. Mol Cancer 2024; 23:9. [PMID: 38195537 PMCID: PMC10775503 DOI: 10.1186/s12943-023-01925-5] [Citation(s) in RCA: 49] [Impact Index Per Article: 49.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 12/20/2023] [Indexed: 01/11/2024] Open
Abstract
The CRISPR system is a revolutionary genome editing tool that has the potential to revolutionize the field of cancer research and therapy. The ability to precisely target and edit specific genetic mutations that drive the growth and spread of tumors has opened up new possibilities for the development of more effective and personalized cancer treatments. In this review, we will discuss the different CRISPR-based strategies that have been proposed for cancer therapy, including inactivating genes that drive tumor growth, enhancing the immune response to cancer cells, repairing genetic mutations that cause cancer, and delivering cancer-killing molecules directly to tumor cells. We will also summarize the current state of preclinical studies and clinical trials of CRISPR-based cancer therapy, highlighting the most promising results and the challenges that still need to be overcome. Safety and delivery are also important challenges for CRISPR-based cancer therapy to become a viable clinical option. We will discuss the challenges and limitations that need to be overcome, such as off-target effects, safety, and delivery to the tumor site. Finally, we will provide an overview of the current challenges and opportunities in the field of CRISPR-based cancer therapy and discuss future directions for research and development. The CRISPR system has the potential to change the landscape of cancer research, and this review aims to provide an overview of the current state of the field and the challenges that need to be overcome to realize this potential.
Collapse
Affiliation(s)
- Mohammad Chehelgerdi
- Novin Genome (NG) Lab, Research and Development Center for Biotechnology, Shahrekord, Iran.
- Young Researchers and Elite Club, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran.
| | - Matin Chehelgerdi
- Novin Genome (NG) Lab, Research and Development Center for Biotechnology, Shahrekord, Iran
- Young Researchers and Elite Club, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | - Milad Khorramian-Ghahfarokhi
- Division of Biotechnology, Department of Pathobiology, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | | | - Esmaeil Mahmoudi
- Young Researchers and Elite Club, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | - Fatemeh Eskandari
- Faculty of Molecular and Cellular Biology -Genetics, Islamic Azad University of Falavarjan, Isfahan, Iran
| | - Mohsen Rashidi
- Department Pharmacology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
- The Health of Plant and Livestock Products Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| | - Asghar Arshi
- Young Researchers and Elite Club, Najafabad Branch, Islamic Azad University, Najafabad, Iran
| | - Abbas Mokhtari-Farsani
- Novin Genome (NG) Lab, Research and Development Center for Biotechnology, Shahrekord, Iran
- Department of Biology, Nourdanesh Institute of Higher Education, Meymeh, Isfahan, Iran
| |
Collapse
|
17
|
Peters IJA, de Pater E, Zhang W. The role of GATA2 in adult hematopoiesis and cell fate determination. Front Cell Dev Biol 2023; 11:1250827. [PMID: 38033856 PMCID: PMC10682726 DOI: 10.3389/fcell.2023.1250827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 10/31/2023] [Indexed: 12/02/2023] Open
Abstract
The correct maintenance and differentiation of hematopoietic stem cells (HSC) in bone marrow is vital for the maintenance and operation of the human blood system. GATA2 plays a critical role in the maintenance of HSCs and the specification of HSCs into the different hematopoietic lineages, highlighted by the various defects observed in patients with heterozygous mutations in GATA2, resulting in cytopenias, bone marrow failure and increased chance of myeloid malignancy, termed GATA2 deficiency syndrome. Despite this, the mechanisms underlying GATA2 deficiency syndrome remain to be elucidated. The detailed description of how GATA2 regulates HSC maintenance and blood lineage determination is crucial to unravel the pathogenesis of GATA2 deficiency syndrome. In this review, we summarize current advances in elucidating the role of GATA2 in hematopoietic cell fate determination and discuss the challenges of modeling GATA2 deficiency syndrome.
Collapse
Affiliation(s)
| | | | - Wei Zhang
- *Correspondence: Wei Zhang, ; Emma de Pater,
| |
Collapse
|
18
|
Rochford AE, Carnicer-Lombarte A, Kawan M, Jin A, Hilton S, Curto VF, Rutz AL, Moreau T, Kotter MR, Malliaras GG, Barone DG. Functional neurological restoration of amputated peripheral nerve using biohybrid regenerative bioelectronics. SCIENCE ADVANCES 2023; 9:eadd8162. [PMID: 36947608 PMCID: PMC10032597 DOI: 10.1126/sciadv.add8162] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 02/16/2023] [Indexed: 06/18/2023]
Abstract
The development of neural interfaces with superior biocompatibility and improved tissue integration is vital for treating and restoring neurological functions in the nervous system. A critical factor is to increase the resolution for mapping neuronal inputs onto implants. For this purpose, we have developed a new category of neural interface comprising induced pluripotent stem cell (iPSC)-derived myocytes as biological targets for peripheral nerve inputs that are grafted onto a flexible electrode arrays. We show long-term survival and functional integration of a biohybrid device carrying human iPSC-derived cells with the forearm nerve bundle of freely moving rats, following 4 weeks of implantation. By improving the tissue-electronics interface with an intermediate cell layer, we have demonstrated enhanced resolution and electrical recording in vivo as a first step toward restorative therapies using regenerative bioelectronics.
Collapse
Affiliation(s)
- Amy E. Rochford
- Electrical Engineering Division, Department of Engineering, University of Cambridge, Cambridge, UK
| | | | - Malak Kawan
- Electrical Engineering Division, Department of Engineering, University of Cambridge, Cambridge, UK
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Amy Jin
- Electrical Engineering Division, Department of Engineering, University of Cambridge, Cambridge, UK
| | - Sam Hilton
- Electrical Engineering Division, Department of Engineering, University of Cambridge, Cambridge, UK
| | - Vincenzo F. Curto
- Electrical Engineering Division, Department of Engineering, University of Cambridge, Cambridge, UK
| | - Alexandra L. Rutz
- Electrical Engineering Division, Department of Engineering, University of Cambridge, Cambridge, UK
| | | | - Mark R. N. Kotter
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- Bit Bio, Cambridge, UK
| | - George G. Malliaras
- Electrical Engineering Division, Department of Engineering, University of Cambridge, Cambridge, UK
| | - Damiano G. Barone
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| |
Collapse
|
19
|
Haque M, Boardman DA, Lam AJ, MacDonald KN, Sanderink L, Huang Q, Fung VCW, Ivison S, Mojibian M, Levings MK. Modelling Graft-Versus-Host Disease in Mice Using Human Peripheral Blood Mononuclear Cells. Bio Protoc 2022; 12:e4566. [PMID: 36561116 PMCID: PMC9729982 DOI: 10.21769/bioprotoc.4566] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 09/12/2022] [Accepted: 10/17/2022] [Indexed: 12/12/2022] Open
Abstract
Graft-versus-host disease (GvHD) is a significant complication of allogeneic hematopoietic stem cell transplantation. In order to develop new therapeutic approaches, there is a need to recapitulate GvHD effects in pre-clinical, in vivo systems, such as mouse and humanized mouse models. In humanized mouse models of GvHD, mice are reconstituted with human immune cells, which become activated by xenogeneic (xeno) stimuli, causing a multi-system disorder known as xenoGvHD. Testing the ability of new therapies to prevent or delay the development of xenoGvHD is often used as pre-clinical, proof-of-concept data, creating the need for standardized methodology to induce, monitor, and report xenoGvHD. Here, we describe detailed methods for how to induce xenoGvHD by injecting human peripheral blood mononuclear cells into immunodeficient NOD SCID gamma mice. We provide comprehensive details on methods for human T cell preparation and injection, mouse monitoring, data collection, interpretation, and reporting. Additionally, we provide an example of the potential utility of the xenoGvHD model to assess the biological activity of a regulatory T-cell therapy. Use of this protocol will allow better standardization of this model and comparison of datasets across different studies. Graft-versus-host disease (GvHD) is a significant complication of allogeneic hematopoietic stem cell transplantation. In order to develop new therapeutic approaches, there is a need to recapitulate GvHD effects in pre-clinical, in vivo systems, such as mouse and humanized mouse models. In humanized mouse models of GvHD, mice are reconstituted with human immune cells, which become activated by xenogeneic (xeno) stimuli, causing a multi-system disorder known as xenoGvHD. Testing the ability of new therapies to prevent or delay the development of xenoGvHD is often used as pre-clinical, proof-of-concept data, creating the need for standardized methodology to induce, monitor, and report xenoGvHD. Here, we describe detailed methods for how to induce xenoGvHD by injecting human peripheral blood mononuclear cells into immunodeficient NOD SCID gamma mice. We provide comprehensive details on methods for human T cell preparation and injection, mouse monitoring, data collection, interpretation, and reporting. Additionally, we provide an example of the potential utility of the xenoGvHD model to assess the biological activity of a regulatory T-cell therapy. Use of this protocol will allow better standardization of this model and comparison of datasets across different studies. This protocol was validated in: Sci Transl Med (2020), DOI: 10.1126/scitranslmed.aaz3866 Graphical abstract.
Collapse
Affiliation(s)
- Manjurul Haque
- Department of Surgery, The University of British Columbia, Vancouver BC, Canada
,
BC Children’s Hospital Research Institute, Vancouver BC, Canada
| | - Dominic A. Boardman
- Department of Surgery, The University of British Columbia, Vancouver BC, Canada
,
BC Children’s Hospital Research Institute, Vancouver BC, Canada
| | - Avery J. Lam
- Department of Surgery, The University of British Columbia, Vancouver BC, Canada
,
BC Children’s Hospital Research Institute, Vancouver BC, Canada
| | - Katherine N. MacDonald
- BC Children’s Hospital Research Institute, Vancouver BC, Canada
,
School of Biomedical Engineering, The University of British Columbia, Vancouver BC, Canada
| | - Lieke Sanderink
- Department of Surgery, The University of British Columbia, Vancouver BC, Canada
,
BC Children’s Hospital Research Institute, Vancouver BC, Canada
| | - Qing Huang
- Department of Surgery, The University of British Columbia, Vancouver BC, Canada
,
BC Children’s Hospital Research Institute, Vancouver BC, Canada
| | - Vivian C. W. Fung
- Department of Surgery, The University of British Columbia, Vancouver BC, Canada
,
BC Children’s Hospital Research Institute, Vancouver BC, Canada
| | - Sabine Ivison
- Department of Surgery, The University of British Columbia, Vancouver BC, Canada
,
BC Children’s Hospital Research Institute, Vancouver BC, Canada
| | - Majid Mojibian
- Department of Surgery, The University of British Columbia, Vancouver BC, Canada
,
BC Children’s Hospital Research Institute, Vancouver BC, Canada
| | - Megan K. Levings
- Department of Surgery, The University of British Columbia, Vancouver BC, Canada
,
BC Children’s Hospital Research Institute, Vancouver BC, Canada
,
School of Biomedical Engineering, The University of British Columbia, Vancouver BC, Canada
,
*For correspondence:
| |
Collapse
|
20
|
Sitta J, Claudio PP, Howard CM. Virus-Based Immuno-Oncology Models. Biomedicines 2022; 10:biomedicines10061441. [PMID: 35740462 PMCID: PMC9220907 DOI: 10.3390/biomedicines10061441] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 06/04/2022] [Accepted: 06/15/2022] [Indexed: 12/12/2022] Open
Abstract
Immunotherapy has been extensively explored in recent years with encouraging results in selected types of cancer. Such success aroused interest in the expansion of such indications, requiring a deep understanding of the complex role of the immune system in carcinogenesis. The definition of hot vs. cold tumors and the role of the tumor microenvironment enlightened the once obscure understanding of low response rates of solid tumors to immune check point inhibitors. Although the major scope found in the literature focuses on the T cell modulation, the innate immune system is also a promising oncolytic tool. The unveiling of the tumor immunosuppressive pathways, lead to the development of combined targeted therapies in an attempt to increase immune infiltration capability. Oncolytic viruses have been explored in different scenarios, in combination with various chemotherapeutic drugs and, more recently, with immune check point inhibitors. Moreover, oncolytic viruses may be engineered to express tumor specific pro-inflammatory cytokines, antibodies, and antigens to enhance immunologic response or block immunosuppressive mechanisms. Development of preclinical models capable to replicate the human immunologic response is one of the major challenges faced by these studies. A thorough understanding of immunotherapy and oncolytic viruses’ mechanics is paramount to develop reliable preclinical models with higher chances of successful clinical therapy application. Thus, in this article, we review current concepts in cancer immunotherapy including the inherent and synthetic mechanisms of immunologic enhancement utilizing oncolytic viruses, immune targeting, and available preclinical animal models, their advantages, and limitations.
Collapse
Affiliation(s)
- Juliana Sitta
- Department of Radiology, University of Mississippi Medical Center, Jackson, MS 39216, USA;
| | - Pier Paolo Claudio
- Department of BioMolecular Sciences, Department of Radiation Oncology, Cancer Center & Research Institute, University of Mississippi Medical Center, Jackson, MS 39216, USA;
| | - Candace M. Howard
- Department of Radiology, University of Mississippi Medical Center, Jackson, MS 39216, USA;
- Correspondence:
| |
Collapse
|
21
|
Görner S, Heim C, Weigmann B, von Silva-Tarouca B, Kuckhahn A, Ramsperger-Gleixner M, Zimmermann R, Weyand M, Ensminger SM. Direct Impact of Human Platelets on the Development of Transplant Arteriosclerosis. Transplantation 2022; 106:1180-1192. [PMID: 34468430 DOI: 10.1097/tp.0000000000003935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Platelets play an important role in the pathogenesis of inflammatory and proliferative vascular changes. The aim of this study was to investigate whether human platelets are able to induce transplant arteriosclerosis in a humanized C57/Bl6-Rag2-/-γc-/- mouse xenograft model. METHODS Nonactivated and in vitro-activated human platelets were analyzed and phenotyped for surface markers by flow cytometry. Side branches of human mammary arteries were implanted into the infrarenal aorta of recipients, followed by daily application of human platelets and histological analyzed on day 30 after transplantation. RESULTS Human platelets collected by apheresis had low levels of platelet activation markers. However, after in vitro activation, expression was markedly increased. Sixty minutes after injection in recipient mice, nonactivated human platelets become significantly activated. Increased adhesion of platelets to the vascular endothelium was detected by in vivo fluorescence microscopy. After intravenous injection of nonactivated or activated platelets, human xenografts showed pronounced intimal proliferation. Immunohistological analysis showed that the group treated with activated human platelets exhibited significantly increased intragraft protein expression of intracellular adhesion molecule-1 and platelet-derived growth factor receptor beta and smooth muscle cell migration into the neointima. CONCLUSIONS These data demonstrate that an isolated daily application of both in vivo- and in vitro-activated human platelets results in the development of transplant arteriosclerosis in a humanized mouse transplantation model.
Collapse
Affiliation(s)
- Susann Görner
- Department of Cardiac Surgery, Friedrich-Alexander University, Erlangen-Nürnberg, Germany
| | - Christian Heim
- Department of Cardiac Surgery, Friedrich-Alexander University, Erlangen-Nürnberg, Germany
| | - Benno Weigmann
- Department of Medicine 1, Friedrich-Alexander University, Erlangen-Nürnberg, Germany
| | | | - Annika Kuckhahn
- Department of Cardiac Surgery, Friedrich-Alexander University, Erlangen-Nürnberg, Germany
| | | | - Robert Zimmermann
- Department of Transfusion Medicine, Friedrich-Alexander University, Erlangen-Nürnberg, Germany
| | - Michael Weyand
- Department of Cardiac Surgery, Friedrich-Alexander University, Erlangen-Nürnberg, Germany
| | - Stephan M Ensminger
- Department of Cardiac Surgery, Friedrich-Alexander University, Erlangen-Nürnberg, Germany
- Present address: Department of Cardiac and Thoracic Vascular Surgery, University Heart Center Lübeck, University Hospital Schleswig Holstein, Lübeck, Germany
| |
Collapse
|
22
|
Song Z, Shao W, Song L, Pei X, Li C. Human Hepatocyte Transduction with Adeno-Associated Virus Vector. Methods Mol Biol 2022; 2544:83-93. [PMID: 36125711 DOI: 10.1007/978-1-0716-2557-6_5] [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: 06/15/2023]
Abstract
As the adeno-associated virus (AAV) vectors hold unique advantages over other viral vectors, AAV gene therapy has accumulated rapid progress and development. Liver-targeted gene therapy by AAV vectors has been successfully applied in clinical trials for many diseases. Low transduction efficiency and high prevalence of neutralizing antibodies (Nabs), however, are the major obstacles to further translate this therapeutic strategy into clinical trials. Pre-clinical evaluation on hepatocytes could help to elucidate the tropism of AAV serotypes for liver-targeted gene therapy, and could also provide a test model to develop novel AAV mutants with Nabs evasion and high liver tropism. Here, we described the basic laboratory procedure to apply the AAV vector to transduce human hepatocytes in vitro and in vivo with some tips gained from our own experience.
Collapse
Affiliation(s)
- Zhenwei Song
- Gene Therapy Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Wenwei Shao
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
| | - Liujiang Song
- Gene Therapy Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Xieolei Pei
- Gene Therapy Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Chengwen Li
- Gene Therapy Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
23
|
Zbinden A, Canté-Barrett K, Pike-Overzet K, Staal FJT. Stem Cell-Based Disease Models for Inborn Errors of Immunity. Cells 2021; 11:cells11010108. [PMID: 35011669 PMCID: PMC8750661 DOI: 10.3390/cells11010108] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 12/23/2021] [Accepted: 12/29/2021] [Indexed: 11/24/2022] Open
Abstract
The intrinsic capacity of human hematopoietic stem cells (hHSCs) to reconstitute myeloid and lymphoid lineages combined with their self-renewal capacity hold enormous promises for gene therapy as a viable treatment option for a number of immune-mediated diseases, most prominently for inborn errors of immunity (IEI). The current development of such therapies relies on disease models, both in vitro and in vivo, which allow the study of human pathophysiology in great detail. Here, we discuss the current challenges with regards to developmental origin, heterogeneity and the subsequent implications for disease modeling. We review models based on induced pluripotent stem cell technology and those relaying on use of adult hHSCs. We critically review the advantages and limitations of current models for IEI both in vitro and in vivo. We conclude that existing and future stem cell-based models are necessary tools for developing next generation therapies for IEI.
Collapse
|
24
|
Jarvis LB, Rainbow DB, Coppard V, Howlett SK, Georgieva Z, Davies JL, Mullay HK, Hester J, Ashmore T, Van Den Bosch A, Grist JT, Coles AJ, Mousa HS, Pluchino S, Mahbubani KT, Griffin JL, Saeb-Parsy K, Issa F, Peruzzotti-Jametti L, Wicker LS, Jones JL. Therapeutically expanded human regulatory T-cells are super-suppressive due to HIF1A induced expression of CD73. Commun Biol 2021; 4:1186. [PMID: 34650224 PMCID: PMC8516976 DOI: 10.1038/s42003-021-02721-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 09/27/2021] [Indexed: 12/14/2022] Open
Abstract
The adoptive transfer of regulatory T-cells (Tregs) is a promising therapeutic approach in transplantation and autoimmunity. However, because large cell numbers are needed to achieve a therapeutic effect, in vitro expansion is required. By comparing their function, phenotype and transcriptomic profile against ex vivo Tregs, we demonstrate that expanded human Tregs switch their metabolism to aerobic glycolysis and show enhanced suppressive function through hypoxia-inducible factor 1-alpha (HIF1A) driven acquisition of CD73 expression. In conjunction with CD39, CD73 expression enables expanded Tregs to convert ATP to immunosuppressive adenosine. We conclude that for maximum therapeutic benefit, Treg expansion protocols should be optimised for CD39/CD73 co-expression.
Collapse
Affiliation(s)
- Lorna B Jarvis
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Daniel B Rainbow
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- JDRF/Wellcome Diabetes and Inflammation Laboratory, Wellcome Centre for Human Genetics, Nuffield Department of Medicine, NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Valerie Coppard
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Sarah K Howlett
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Zoya Georgieva
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Jessica L Davies
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | | | - Joanna Hester
- Department of Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Tom Ashmore
- Department of Biochemistry and Cambridge Systems Biology Centre, University of Cambridge, Cambridge, UK
| | | | - James T Grist
- Department of Radiology, University of Cambridge, Cambridge, UK
| | - Alasdair J Coles
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Hani S Mousa
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Stefano Pluchino
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | | | - Julian L Griffin
- Department of Biochemistry and Cambridge Systems Biology Centre, University of Cambridge, Cambridge, UK
- Imperial College London Dementia Research Institute & Section of Biomolecular Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | | | - Fadi Issa
- Department of Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | | | - Linda S Wicker
- JDRF/Wellcome Diabetes and Inflammation Laboratory, Wellcome Centre for Human Genetics, Nuffield Department of Medicine, NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Joanne L Jones
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK.
| |
Collapse
|
25
|
Petrus-Reurer S, Romano M, Howlett S, Jones JL, Lombardi G, Saeb-Parsy K. Immunological considerations and challenges for regenerative cellular therapies. Commun Biol 2021; 4:798. [PMID: 34172826 PMCID: PMC8233383 DOI: 10.1038/s42003-021-02237-4] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 05/17/2021] [Indexed: 02/06/2023] Open
Abstract
The central goal of regenerative medicine is to replace damaged or diseased tissue with cells that integrate and function optimally. The capacity of pluripotent stem cells to produce unlimited numbers of differentiated cells is of considerable therapeutic interest, with several clinical trials underway. However, the host immune response represents an important barrier to clinical translation. Here we describe the role of the host innate and adaptive immune responses as triggers of allogeneic graft rejection. We discuss how the immune response is determined by the cellular therapy. Additionally, we describe the range of available in vitro and in vivo experimental approaches to examine the immunogenicity of cellular therapies, and finally we review potential strategies to ameliorate immune rejection. In conclusion, we advocate establishment of platforms that bring together the multidisciplinary expertise and infrastructure necessary to comprehensively investigate the immunogenicity of cellular therapies to ensure their clinical safety and efficacy.
Collapse
Affiliation(s)
- Sandra Petrus-Reurer
- Department of Surgery, University of Cambridge, and NIHR Cambridge Biomedical Research Centre, Cambridge, United Kingdom.
| | - Marco Romano
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, Guy's Hospital, London, United Kingdom
| | - Sarah Howlett
- Department of Clinical Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Joanne Louise Jones
- Department of Clinical Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Giovanna Lombardi
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, Guy's Hospital, London, United Kingdom
| | - Kourosh Saeb-Parsy
- Department of Surgery, University of Cambridge, and NIHR Cambridge Biomedical Research Centre, Cambridge, United Kingdom.
| |
Collapse
|
26
|
Chenouard V, Remy S, Tesson L, Ménoret S, Ouisse LH, Cherifi Y, Anegon I. Advances in Genome Editing and Application to the Generation of Genetically Modified Rat Models. Front Genet 2021; 12:615491. [PMID: 33959146 PMCID: PMC8093876 DOI: 10.3389/fgene.2021.615491] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 02/22/2021] [Indexed: 12/13/2022] Open
Abstract
The rat has been extensively used as a small animal model. Many genetically engineered rat models have emerged in the last two decades, and the advent of gene-specific nucleases has accelerated their generation in recent years. This review covers the techniques and advances used to generate genetically engineered rat lines and their application to the development of rat models more broadly, such as conditional knockouts and reporter gene strains. In addition, genome-editing techniques that remain to be explored in the rat are discussed. The review also focuses more particularly on two areas in which extensive work has been done: human genetic diseases and immune system analysis. Models are thoroughly described in these two areas and highlight the competitive advantages of rat models over available corresponding mouse versions. The objective of this review is to provide a comprehensive description of the advantages and potential of rat models for addressing specific scientific questions and to characterize the best genome-engineering tools for developing new projects.
Collapse
Affiliation(s)
- Vanessa Chenouard
- CHU Nantes, Inserm, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, Université de Nantes, Nantes, France
- genOway, Lyon, France
| | - Séverine Remy
- CHU Nantes, Inserm, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, Université de Nantes, Nantes, France
| | - Laurent Tesson
- CHU Nantes, Inserm, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, Université de Nantes, Nantes, France
| | - Séverine Ménoret
- CHU Nantes, Inserm, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, Université de Nantes, Nantes, France
- CHU Nantes, Inserm, CNRS, SFR Santé, Inserm UMS 016, CNRS UMS 3556, Nantes Université, Nantes, France
| | - Laure-Hélène Ouisse
- CHU Nantes, Inserm, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, Université de Nantes, Nantes, France
| | | | - Ignacio Anegon
- CHU Nantes, Inserm, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, Université de Nantes, Nantes, France
| |
Collapse
|
27
|
Adigbli G, Hua P, Uchiyama M, Roberts I, Hester J, Watt SM, Issa F. Development of LT-HSC-Reconstituted Non-Irradiated NBSGW Mice for the Study of Human Hematopoiesis In Vivo. Front Immunol 2021; 12:642198. [PMID: 33868276 PMCID: PMC8044770 DOI: 10.3389/fimmu.2021.642198] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 03/03/2021] [Indexed: 11/26/2022] Open
Abstract
Humanized immune system (HIS) mouse models are useful tools for the in vivo investigation of human hematopoiesis. However, the majority of HIS models currently in use are biased towards lymphocyte development and fail to support long-term multilineage leucocytes and erythrocytes. Those that achieve successful multilineage reconstitution often require preconditioning steps which are expensive, cause animal morbidity, are technically demanding, and poorly reproducible. In this study, we address this challenge by using HSPC-NBSGW mice, in which NOD,B6.SCID IL-2rγ-/-KitW41/W41 (NBSGW) mice are engrafted with human CD133+ hematopoietic stem and progenitor cells (HSPCs) without the need for preconditioning by sublethal irradiation. These HSPCs are enriched in long-term hematopoietic stem cells (LT-HSCs), while NBSGW mice are permissive to human hematopoietic stem cell (HSC) engraftment, thus reducing the cell number required for successful HIS development. B cells reconstitute with the greatest efficiency, including mature B cells capable of class-switching following allogeneic stimulation and, within lymphoid organs and peripheral blood, T cells at a spectrum of stages of maturation. In the thymus, human thymocytes are identified at all major stages of development. Phenotypically distinct subsets of myeloid cells, including dendritic cells and mature monocytes, engraft to a variable degree in the bone marrow and spleen, and circulate in peripheral blood. Finally, we observe human erythrocytes which persist in the periphery at high levels following macrophage clearance. The HSPC-NBSGW model therefore provides a useful platform for the study of human hematological and immunological processes and pathologies.
Collapse
Affiliation(s)
- George Adigbli
- Transplantation Research and Immunology Group, John Radcliffe Hospital, Nuffield Department of Surgical Sciences, University of Oxford, Oxford, United Kingdom
| | - Peng Hua
- MRC Molecular Haematology Unit, Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, Oxford, United Kingdom
- Nuffield Division of Clinical Laboratory Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Masateru Uchiyama
- Transplantation Research and Immunology Group, John Radcliffe Hospital, Nuffield Department of Surgical Sciences, University of Oxford, Oxford, United Kingdom
| | - Irene Roberts
- MRC Molecular Haematology Unit, Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, Oxford, United Kingdom
- Department of Paediatrics, Children’s Hospital, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Joanna Hester
- Transplantation Research and Immunology Group, John Radcliffe Hospital, Nuffield Department of Surgical Sciences, University of Oxford, Oxford, United Kingdom
| | - Suzanne M. Watt
- Nuffield Division of Clinical Laboratory Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, and Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| | - Fadi Issa
- Transplantation Research and Immunology Group, John Radcliffe Hospital, Nuffield Department of Surgical Sciences, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
28
|
Vimond N, Lasselin J, Anegon I, Guillonneau C, Bézie S. Genetic engineering of human and mouse CD4 + and CD8 + Tregs using lentiviral vectors encoding chimeric antigen receptors. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2021; 20:69-85. [PMID: 33376756 PMCID: PMC7749301 DOI: 10.1016/j.omtm.2020.11.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 11/10/2020] [Indexed: 11/30/2022]
Abstract
The last decade has seen a significant increase of cell therapy protocols using effector T cells (Teffs) in particular, but also, more recently, non-engineered and expanded polyclonal regulatory T cells (Tregs) to control pathological immune responses such as cancer, autoimmune diseases, or transplantation rejection. However, limitations, such as stability, migration, and specificity of the cell products, have been seen. Thus, genetic engineering of these cell subsets is expected to provide the next generation of T cell therapy products. Lentiviral vectors are commonly used to modify Teffs; however, Tregs are more sensitive to mechanical stress and require specific culture conditions. Also, there is a lack of reproducible and efficient protocols to expand and genetically modify Tregs without affecting their growth and function. Due to smaller number of cells and poorer viability upon culture in vitro, mouse Tregs are more difficult to transduce and amplify in vitro than human Tregs. Here we propose a step-by-step protocol to produce both human and mouse genetically modified CD8+ and CD4+ Tregs in sufficient amounts to assess their therapeutic efficacy in humanized immunocompromised mouse models and murine models of disease and to establish pre-clinical proofs of concept. We report, for the first time, an efficient and reproducible method to isolate Tregs from human blood or mouse spleen, transduce with a lentiviral vector, and culture, in parallel, CD8+ and CD4+ Tregs while preserving their function. Beyond chimeric antigen receptor (CAR)-Treg cell therapy, this protocol will promote the development of potential new engineered T cell therapies to treat autoimmune diseases and transplantation rejection.
Collapse
Affiliation(s)
- Nadège Vimond
- Université de Nantes, CHU Nantes, Inserm, CNRS, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, 44093 Nantes Cedex 01, France
| | - Juliette Lasselin
- Université de Nantes, CHU Nantes, Inserm, CNRS, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, 44093 Nantes Cedex 01, France
| | - Ignacio Anegon
- Université de Nantes, CHU Nantes, Inserm, CNRS, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, 44093 Nantes Cedex 01, France
| | - Carole Guillonneau
- Université de Nantes, CHU Nantes, Inserm, CNRS, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, 44093 Nantes Cedex 01, France
- Corresponding author: Carole Guillonneau, Université de Nantes, CHU Nantes, Inserm, CNRS, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, 30 Bd Jean Monnet, 44093, Nantes Cedex 01, France.
| | - Séverine Bézie
- Université de Nantes, CHU Nantes, Inserm, CNRS, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, 44093 Nantes Cedex 01, France
- Corresponding author: Séverine Bézie, Université de Nantes, CHU Nantes, Inserm, CNRS, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, 30 Bd Jean Monnet, 44093, Nantes Cedex 01, France.
| |
Collapse
|
29
|
Amini L, Greig J, Schmueck-Henneresse M, Volk HD, Bézie S, Reinke P, Guillonneau C, Wagner DL, Anegon I. Super-Treg: Toward a New Era of Adoptive Treg Therapy Enabled by Genetic Modifications. Front Immunol 2021; 11:611638. [PMID: 33717052 PMCID: PMC7945682 DOI: 10.3389/fimmu.2020.611638] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 12/24/2020] [Indexed: 12/27/2022] Open
Abstract
Regulatory Tcells (Treg) are essential components of peripheral immune homeostasis. Adoptive Treg cell therapy has shown efficacy in a variety of immune-mediated diseases in preclinical studies and is now moving from phase I/IIa to larger phase II studies aiming to demonstrate efficacy. However, hurdles such as in vivo stability and efficacy remain to be addressed. Nevertheless, preclinical models have shown that Treg function and specificity can be increased by pharmacological substances or gene modifications, and even that conventional T cells can be converted to Treg potentially providing new sources of Treg and facilitating Treg cell therapy. The exponential growth in genetic engineering techniques and their application to T cells coupled to a large body of knowledge on Treg open numerous opportunities to generate Treg with "superpowers". This review summarizes the genetic engineering techniques available and their applications for the next-generation of Super-Treg with increased function, stability, redirected specificity and survival.
Collapse
Affiliation(s)
- Leila Amini
- BIH Center for Regenerative Therapies (BCRT) and Berlin Center for Advanced Therapies (BeCAT), Charité-Universitätsmedizin Berlin and Berlin Institute of Health (BIH), Berlin, Germany
| | - Jenny Greig
- INSERM, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, Nantes, France
| | - Michael Schmueck-Henneresse
- BIH Center for Regenerative Therapies (BCRT) and Berlin Center for Advanced Therapies (BeCAT), Charité-Universitätsmedizin Berlin and Berlin Institute of Health (BIH), Berlin, Germany
| | - Hans-Dieter Volk
- BIH Center for Regenerative Therapies (BCRT) and Berlin Center for Advanced Therapies (BeCAT), Charité-Universitätsmedizin Berlin and Berlin Institute of Health (BIH), Berlin, Germany
| | - Séverine Bézie
- INSERM, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, Nantes, France
| | - Petra Reinke
- BIH Center for Regenerative Therapies (BCRT) and Berlin Center for Advanced Therapies (BeCAT), Charité-Universitätsmedizin Berlin and Berlin Institute of Health (BIH), Berlin, Germany
| | - Carole Guillonneau
- INSERM, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, Nantes, France
| | - Dimitrios L. Wagner
- BIH Center for Regenerative Therapies (BCRT) and Berlin Center for Advanced Therapies (BeCAT), Charité-Universitätsmedizin Berlin and Berlin Institute of Health (BIH), Berlin, Germany
| | - Ignacio Anegon
- INSERM, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, Nantes, France
| |
Collapse
|
30
|
Maser IP, Hoves S, Bayer C, Heidkamp G, Nimmerjahn F, Eckmann J, Ries CH. The Tumor Milieu Promotes Functional Human Tumor-Resident Plasmacytoid Dendritic Cells in Humanized Mouse Models. Front Immunol 2020; 11:2082. [PMID: 33013879 PMCID: PMC7507800 DOI: 10.3389/fimmu.2020.02082] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 07/30/2020] [Indexed: 12/11/2022] Open
Abstract
Particular interest to harness the innate immune system for cancer immunotherapy is fueled by limitations of immune checkpoint blockade. Plasmacytoid dendritic cells (pDC) are detected in a variety of solid tumors and correlate with poor clinical outcome. Release of type I interferons in response to toll-like-receptor (TLR)7 and TLR9 activation is the pDC hallmark. Mouse and human pDC differ substantially in their biology concerning surface marker expression and cytokine production. Here, we employed humanized mouse models (HIS) to study pDC function. We performed a comprehensive characterization of transgenic, myeloid-enhanced mouse strains (NOG-EXL and NSG-SGM3) expressing human interleukin-3 (hIL-3) and granulocyte-macrophage colony stimulating factor (GM-CSF) using identical humanization protocols. Only in HIS-NOG-EXL mice sufficient pDC infiltration was detectable. Therefore, we selected this strain for subsequent tumor studies. We analyzed pDC frequency in peripheral blood and tumors by comparing HIS-NOG-EXL with HIS-NOG mice bearing three different ovarian and breast tumors. Despite the substantially increased pDC numbers in peripheral blood of HIS-NOG-EXL mice, we detected TLR7/8 agonist responsive and thus functional pDCs only in certain tumor models independent of the mouse strain employed. However, HIS-NOG-EXL mice showed in general a superior humanization phenotype characterized by reconstitution of different myeloid subsets, NK cells and B cells producing physiologic IgG levels. Hence, we provide first evidence that the tumor milieu but not genetically introduced cytokines defines intratumoral (i.t.) frequencies of the rare pDC subset. This study provides model systems to investigate in vivo pro- and anti-tumoral human pDC functions.
Collapse
Affiliation(s)
- Ilona-Petra Maser
- Roche Pharma Research and Early Development, Discovery Oncology, Roche Innovation Center Munich, Penzberg, Germany
| | - Sabine Hoves
- Roche Pharma Research and Early Development, Discovery Oncology, Roche Innovation Center Munich, Penzberg, Germany
| | - Christa Bayer
- Roche Pharma Research and Early Development, Discovery Oncology, Roche Innovation Center Munich, Penzberg, Germany
| | - Gordon Heidkamp
- Roche Pharma Research and Early Development, Discovery Oncology, Roche Innovation Center Munich, Penzberg, Germany
| | - Falk Nimmerjahn
- FAU Erlangen, Division of Genetics, Department of Biology, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Jan Eckmann
- Roche Pharma Research and Early Development, Discovery Oncology, Roche Innovation Center Munich, Penzberg, Germany
| | - Carola H Ries
- Roche Pharma Research and Early Development, Discovery Oncology, Roche Innovation Center Munich, Penzberg, Germany.,Dr. Carola Ries Consulting, Penzberg, Germany
| |
Collapse
|