1
|
Han R, Su L, Cheng L. Advancing Human Vaccine Development Using Humanized Mouse Models. Vaccines (Basel) 2024; 12:1012. [PMID: 39340042 PMCID: PMC11436046 DOI: 10.3390/vaccines12091012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 08/11/2024] [Accepted: 08/27/2024] [Indexed: 09/30/2024] Open
Abstract
The development of effective vaccines against infectious diseases remains a critical challenge in global health. Animal models play a crucial role in vaccine development by providing valuable insights into the efficacy, safety, and mechanisms of immune response induction, which guide the design and formulation of vaccines. However, traditional animal models often inadequately recapitulate human immune responses. Humanized mice (hu-mice) models with a functional human immune system have emerged as invaluable tools in bridging the translational gap between preclinical research and clinical trials for human vaccine development. This review summarizes commonly used hu-mice models and advances in optimizing them to improve human immune responses. We review the application of humanized mice for human vaccine development with a focus on HIV-1 vaccines. We also discuss the remaining challenges and improvements needed for the currently available hu-mice models to better facilitate the development and testing of human vaccines for infectious diseases.
Collapse
Affiliation(s)
- Runpeng Han
- Department of Infectious Diseases, Zhongnan Hospital of Wuhan University, Medical Research Institute, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan 430071, China
- Center for AIDS Research, Wuhan University, Wuhan 430071, China
| | - Lishan Su
- Laboratory of Viral Pathogenesis and Immunotherapy, Institute of Human Virology, Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD 02121, USA
| | - Liang Cheng
- Department of Infectious Diseases, Zhongnan Hospital of Wuhan University, Medical Research Institute, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan 430071, China
- Center for AIDS Research, Wuhan University, Wuhan 430071, China
| |
Collapse
|
2
|
Bin Y, Ren J, Zhang H, Zhang T, Liu P, Xin Z, Yang H, Feng Z, Chen Z, Zhang H. Against all odds: The road to success in the development of human immune reconstitution mice. Animal Model Exp Med 2024; 7:460-470. [PMID: 38591343 PMCID: PMC11369039 DOI: 10.1002/ame2.12407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 03/17/2024] [Indexed: 04/10/2024] Open
Abstract
The mouse genome has a high degree of homology with the human genome, and its physiological, biochemical, and developmental regulation mechanisms are similar to those of humans; therefore, mice are widely used as experimental animals. However, it is undeniable that interspecies differences between humans and mice can lead to experimental errors. The differences in the immune system have become an important factor limiting current immunological research. The application of immunodeficient mice provides a possible solution to these problems. By transplanting human immune cells or tissues, such as peripheral blood mononuclear cells or hematopoietic stem cells, into immunodeficient mice, a human immune system can be reconstituted in the mouse body, and the engrafted immune cells can elicit human-specific immune responses. Researchers have been actively exploring the development and differentiation conditions of host recipient animals and grafts in order to achieve better immune reconstitution. Through genetic engineering methods, immunodeficient mice can be further modified to provide a favorable developmental and differentiation microenvironment for the grafts. From initially only being able to reconstruct single T lymphocyte lineages, it is now possible to reconstruct lymphoid and myeloid cells, providing important research tools for immunology-related studies. In this review, we compare the differences in immune systems of humans and mice, describe the development history of human immune reconstitution from the perspectives of immunodeficient mice and grafts, and discuss the latest advances in enhancing the efficiency of human immune cell reconstitution, aiming to provide important references for immunological related researches.
Collapse
Affiliation(s)
- Yixiao Bin
- School of Basic Medical SciencesShaanxi University of Chinese MedicineXianyangChina
- Department of Cell Biology, National Translational Science Center for Molecular MedicineFourth Military Medical UniversityXi'anChina
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesFourth Military Medical UniversityXi'anChina
| | - Jing Ren
- School of Basic Medical SciencesShaanxi University of Chinese MedicineXianyangChina
- Department of Cell Biology, National Translational Science Center for Molecular MedicineFourth Military Medical UniversityXi'anChina
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesFourth Military Medical UniversityXi'anChina
| | - Haowei Zhang
- Department of Occupational & Environmental Health and the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public HealthFourth Military Medical UniversityXi'anChina
| | - Tianjiao Zhang
- Department of Cell Biology, National Translational Science Center for Molecular MedicineFourth Military Medical UniversityXi'anChina
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesFourth Military Medical UniversityXi'anChina
| | - Peijuan Liu
- Department of Cell Biology, National Translational Science Center for Molecular MedicineFourth Military Medical UniversityXi'anChina
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesFourth Military Medical UniversityXi'anChina
| | - Zhiqian Xin
- Department of Cell Biology, National Translational Science Center for Molecular MedicineFourth Military Medical UniversityXi'anChina
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesFourth Military Medical UniversityXi'anChina
| | - Haijiao Yang
- Department of Cell Biology, National Translational Science Center for Molecular MedicineFourth Military Medical UniversityXi'anChina
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesFourth Military Medical UniversityXi'anChina
| | - Zhuan Feng
- Department of Cell Biology, National Translational Science Center for Molecular MedicineFourth Military Medical UniversityXi'anChina
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesFourth Military Medical UniversityXi'anChina
| | - Zhinan Chen
- Department of Cell Biology, National Translational Science Center for Molecular MedicineFourth Military Medical UniversityXi'anChina
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesFourth Military Medical UniversityXi'anChina
| | - Hai Zhang
- Department of Cell Biology, National Translational Science Center for Molecular MedicineFourth Military Medical UniversityXi'anChina
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesFourth Military Medical UniversityXi'anChina
| |
Collapse
|
3
|
Yu T, Xu Q, Chen X, Deng X, Chen N, Kou MT, Huang Y, Guo J, Xiao Z, Wang J. Biomimetic nanomaterials in myocardial infarction treatment: Harnessing bionic strategies for advanced therapeutics. Mater Today Bio 2024; 25:100957. [PMID: 38322664 PMCID: PMC10844134 DOI: 10.1016/j.mtbio.2024.100957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 01/11/2024] [Accepted: 01/15/2024] [Indexed: 02/08/2024] Open
Abstract
Myocardial infarction (MI) and its associated poor prognosis pose significant risks to human health. Nanomaterials hold great potential for the treatment of MI due to their targeted and controlled release properties, particularly biomimetic nanomaterials. The utilization of biomimetic strategies based on extracellular vesicles (EVs) and cell membranes will serve as the guiding principle for the development of nanomaterial therapy in the future. In this review, we present an overview of research progress on various exosomes derived from mesenchymal stem cells, cardiomyocytes, or induced pluripotent stem cells in the context of myocardial infarction (MI) therapy. These exosomes, utilized as cell-free therapies, have demonstrated the ability to enhance the efficacy of reducing the size of the infarcted area and preventing ischaemic reperfusion through mechanisms such as oxidative stress reduction, polarization modulation, fibrosis inhibition, and angiogenesis promotion. Moreover, EVs can exert cardioprotective effects by encapsulating therapeutic agents and can be engineered to specifically target the infarcted myocardium. Furthermore, we discuss the use of cell membranes derived from erythrocytes, stem cells, immune cells and platelets to encapsulate nanomaterials. This approach allows the nanomaterials to camouflage themselves as endogenous substances targeting the region affected by MI, thereby minimizing toxicity and improving biocompatibility. In conclusion, biomimetic nano-delivery systems hold promise as a potentially beneficial technology for MI treatment. This review serves as a valuable reference for the application of biomimetic nanomaterials in MI therapy and aims to expedite the translation of NPs-based MI therapeutic strategies into practical clinical applications.
Collapse
Affiliation(s)
- Tingting Yu
- Department of Pharmacy, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
- The Guangzhou Key Laboratory of Basic and Translational Research on Chronic Diseases, Jinan University, Guangzhou, 510630, China
| | - Qiaxin Xu
- Department of Pharmacy, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
- The Guangzhou Key Laboratory of Basic and Translational Research on Chronic Diseases, Jinan University, Guangzhou, 510630, China
| | - Xu Chen
- Department of Clinical Pharmacy, Daqing Oilfield General Hospital, Daqing, 163000, China
| | - Xiujiao Deng
- Department of Pharmacy, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
- The Guangzhou Key Laboratory of Basic and Translational Research on Chronic Diseases, Jinan University, Guangzhou, 510630, China
| | - Nenghua Chen
- Department of Pharmacy, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
- The Guangzhou Key Laboratory of Basic and Translational Research on Chronic Diseases, Jinan University, Guangzhou, 510630, China
| | - Man Teng Kou
- Department of Pharmacy, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
- The Guangzhou Key Laboratory of Basic and Translational Research on Chronic Diseases, Jinan University, Guangzhou, 510630, China
| | - Yanyu Huang
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, CA, 95817, USA
| | - Jun Guo
- The Guangzhou Key Laboratory of Basic and Translational Research on Chronic Diseases, Jinan University, Guangzhou, 510630, China
- Department of Cardiology, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Zeyu Xiao
- The Guangzhou Key Laboratory of Basic and Translational Research on Chronic Diseases, Jinan University, Guangzhou, 510630, China
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, Jinan University, Guangzhou, 510630, China
| | - Jinghao Wang
- Department of Pharmacy, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
- The Guangzhou Key Laboratory of Basic and Translational Research on Chronic Diseases, Jinan University, Guangzhou, 510630, China
| |
Collapse
|
4
|
Yang Y, Li J, Li D, Zhou W, Yan F, Wang W. Humanized mouse models: A valuable platform for preclinical evaluation of human cancer. Biotechnol Bioeng 2024; 121:835-852. [PMID: 38151887 DOI: 10.1002/bit.28618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 11/26/2023] [Indexed: 12/29/2023]
Abstract
Animal models are routinely employed to assess the treatments for human cancer. However, due to significant differences in genetic backgrounds, traditional animal models are unable to meet bioresearch needs. To overcome this restriction, researchers have generated and optimized immunodeficient mice, and then engrafted human genes, cells, tissues, or organs in mice so that the responses in the model mice could provide a more reliable reference for treatments. As a bridge connecting clinical application and basic research, humanized mice are increasingly used in the preclinical evaluation of cancer treatments, particularly after gene interleukin 2 receptor gamma mutant mice were generated. Human cancer models established in humanized mice support exploration of the mechanism of cancer occurrence and provide an efficient platform for drug screening. However, it is undeniable that the further application of humanized mice still faces multiple challenges. This review summarizes the construction approaches for humanized mice and their existing limitations. We also report the latest applications of humanized mice in preclinical evaluation for the treatment of cancer and point out directions for future optimization of these models.
Collapse
Affiliation(s)
- Yuening Yang
- State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Jiaqian Li
- State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Dan Li
- State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Weilin Zhou
- State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Feiyang Yan
- State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Wei Wang
- State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
5
|
Kaur S, Roberts DD. Emerging functions of thrombospondin-1 in immunity. Semin Cell Dev Biol 2024; 155:22-31. [PMID: 37258315 PMCID: PMC10684827 DOI: 10.1016/j.semcdb.2023.05.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 05/24/2023] [Indexed: 06/02/2023]
Abstract
Thrombospondin-1 is a secreted matricellular glycoprotein that modulates cell behavior by interacting with components of the extracellular matrix and with several cell surface receptors. Its presence in the extracellular matrix is induced by injuries that cause thrombospondin-1 release from platelets and conditions including hyperglycemia, ischemia, and aging that stimulate its expression by many cell types. Conversely, rapid receptor-mediated clearance of thrombospondin-1 from the extracellular space limits its sustained presence in the extracellular space and maintains sub-nanomolar physiological concentrations in blood plasma. Roles for thrombospondin-1 signaling, mediated by specific cellular receptors or by activation of latent TGFβ, have been defined in T and B lymphocytes, natural killer cells, macrophages, neutrophils, and dendritic cells. In addition to regulating physiological nitric oxide signaling and responses of cells to stress, studies in mice lacking thrombospondin-1 or its receptors have revealed important roles for thrombospondin-1 in regulating immune responses in infectious and autoimmune diseases and antitumor immunity.
Collapse
Affiliation(s)
- Sukhbir Kaur
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - David D Roberts
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
6
|
Du EJ, Muench MO. A Monocytic Barrier to the Humanization of Immunodeficient Mice. Curr Stem Cell Res Ther 2024; 19:959-980. [PMID: 37859310 PMCID: PMC10997744 DOI: 10.2174/011574888x263597231001164351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 08/17/2023] [Accepted: 08/25/2023] [Indexed: 10/21/2023]
Abstract
Mice with severe immunodeficiencies have become very important tools for studying foreign cells in an in vivo environment. Xenotransplants can be used to model cells from many species, although most often, mice are humanized through the transplantation of human cells or tissues to meet the needs of medical research. The development of immunodeficient mice is reviewed leading up to the current state-of-the-art strains, such as the NOD-scid-gamma (NSG) mouse. NSG mice are excellent hosts for human hematopoietic stem cell transplants or immune reconstitution through transfusion of human peripheral blood mononuclear cells. However, barriers to full hematopoietic engraftment still remain; notably, the survival of human cells in the circulation is brief, which limits overall hematological and immune reconstitution. Reports have indicated a critical role for monocytic cells - monocytes, macrophages, and dendritic cells - in the clearance of xenogeneic cells from circulation. Various aspects of the NOD genetic background that affect monocytic cell growth, maturation, and function that are favorable to human cell transplantation are discussed. Important receptors, such as SIRPα, that form a part of the innate immune system and enable the recognition and phagocytosis of foreign cells by monocytic cells are reviewed. The development of humanized mouse models has taken decades of work in creating more immunodeficient mice, genetic modification of these mice to express human genes, and refinement of transplant techniques to optimize engraftment. Future advances may focus on the monocytic cells of the host to find ways for further engraftment and survival of xenogeneic cells.
Collapse
Affiliation(s)
- Emily J. Du
- Vitalant Research Institute, 360 Spear Street, Suite 200, San Francisco, CA, 94105, USA
| | - Marcus O. Muench
- Vitalant Research Institute, 360 Spear Street, Suite 200, San Francisco, CA, 94105, USA
- Department of Laboratory Medicine, University of California, San Francisco, CA, 94143, USA
| |
Collapse
|
7
|
Xia Z, Mu W, Yuan S, Fu S, Liu Y, Zhang N. Cell Membrane Biomimetic Nano-Delivery Systems for Cancer Therapy. Pharmaceutics 2023; 15:2770. [PMID: 38140108 PMCID: PMC10748133 DOI: 10.3390/pharmaceutics15122770] [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: 10/24/2023] [Revised: 11/29/2023] [Accepted: 11/29/2023] [Indexed: 12/24/2023] Open
Abstract
Nano-delivery systems have demonstrated great promise in the therapy of cancer. However, the therapeutic efficacy of conventional nanomedicines is hindered by the clearance of the blood circulation system and the physiological barriers surrounding the tumor. Inspired by the unique capabilities of cells within the body, such as immune evasion, prolonged circulation, and tumor-targeting, there has been a growing interest in developing cell membrane biomimetic nanomedicine delivery systems. Cell membrane modification on nanoparticle surfaces can prolong circulation time, activate tumor-targeting, and ultimately improve the efficacy of cancer treatment. It shows excellent development potential. This review will focus on the advancements in various cell membrane nano-drug delivery systems for cancer therapy and the obstacles encountered during clinical implementation. It is hoped that such discussions will inspire the development of cell membrane biomimetic nanomedical systems.
Collapse
Affiliation(s)
- Zhenxing Xia
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, Jinan 250012, China; (Z.X.); (W.M.); (S.Y.); (S.F.)
- Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan 250012, China
| | - Weiwei Mu
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, Jinan 250012, China; (Z.X.); (W.M.); (S.Y.); (S.F.)
- Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan 250012, China
| | - Shijun Yuan
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, Jinan 250012, China; (Z.X.); (W.M.); (S.Y.); (S.F.)
- Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan 250012, China
| | - Shunli Fu
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, Jinan 250012, China; (Z.X.); (W.M.); (S.Y.); (S.F.)
- Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan 250012, China
| | - Yongjun Liu
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, Jinan 250012, China; (Z.X.); (W.M.); (S.Y.); (S.F.)
- Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan 250012, China
| | - Na Zhang
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, Jinan 250012, China; (Z.X.); (W.M.); (S.Y.); (S.F.)
- Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan 250012, China
| |
Collapse
|
8
|
Thaller AL, Jönsson F, Fiquet O, Marie S, Doisne JM, Girelli-Zubani G, Eri T, Fernandes P, Tatirovsky E, Langa-Vives F, Bruhns P, Strick-Marchand H, Di Santo JP. A human immune system (HIS) mouse model that dissociates roles for mouse and human FcR + cells during antibody-mediated immune responses. Eur J Immunol 2023; 53:e2350454. [PMID: 37621208 DOI: 10.1002/eji.202350454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 07/21/2023] [Accepted: 08/21/2023] [Indexed: 08/26/2023]
Abstract
Human immune system (HIS) mice provide a model to study human immune responses in vivo. Currently available HIS mouse models may harbor mouse Fc Receptor (FcR)-expressing cells that exert potent effector functions following administration of human Ig. Previous studies showed that the ablation of the murine FcR gamma chain (FcR-γ) results in loss of antibody-dependent cellular cytotoxicity and antibody-dependent cellular phagocytosis in vivo. We created a new FcR-γ-deficient HIS mouse model to compare host (mouse) versus graft (human) effects underlying antibody-mediated immune responses in vivo. FcR-γ-deficient HIS recipients lack expression and function of mouse activating FcRs and can be stably and robustly reconstituted with human immune cells. By screening blood B-cell depletion by rituximab Ig variants, we found that human FcγRs-mediated IgG1 effects, whereas mouse activating FcγRs were dominant in IgG4 effects. Complement played a role as an IgG1 variant (IgG1 K322A) lacking complement binding activity was largely ineffective. Finally, we provide evidence that FcγRIIIA on human NK cells could mediate complement-independent B-cell depletion by IgG1 K322A. We anticipate that our FcR-γ-deficient HIS model will help clarify mechanisms of action of exogenous administered human antibodies in vivo.
Collapse
Affiliation(s)
- Anna Louisa Thaller
- Institut Pasteur, Innate Immunity Unit, Université Paris Cité, Inserm U1223, Paris, France
| | - Friederike Jönsson
- Institut Pasteur, Antibodies in Therapy and Pathology Unit, Université Paris Cité, Inserm U1222, Paris, France
| | - Oriane Fiquet
- Institut Pasteur, Innate Immunity Unit, Université Paris Cité, Inserm U1223, Paris, France
| | - Solenne Marie
- Institut Pasteur, Innate Immunity Unit, Université Paris Cité, Inserm U1223, Paris, France
| | - Jean-Marc Doisne
- Institut Pasteur, Innate Immunity Unit, Université Paris Cité, Inserm U1223, Paris, France
| | - Giulia Girelli-Zubani
- Institut Pasteur, Innate Immunity Unit, Université Paris Cité, Inserm U1223, Paris, France
| | - Toshiki Eri
- Institut Pasteur, Innate Immunity Unit, Université Paris Cité, Inserm U1223, Paris, France
| | - Priyanka Fernandes
- Institut Pasteur, Innate Immunity Unit, Université Paris Cité, Inserm U1223, Paris, France
| | - Evgeny Tatirovsky
- Institut Pasteur, Innate Immunity Unit, Université Paris Cité, Inserm U1223, Paris, France
| | - Francina Langa-Vives
- Institut Pasteur, Mouse Genetics Engineering Platform, Université Paris Cité, Paris, France
| | - Pierre Bruhns
- Institut Pasteur, Antibodies in Therapy and Pathology Unit, Université Paris Cité, Inserm U1222, Paris, France
| | - Hélène Strick-Marchand
- Institut Pasteur, Innate Immunity Unit, Université Paris Cité, Inserm U1223, Paris, France
| | - James P Di Santo
- Institut Pasteur, Innate Immunity Unit, Université Paris Cité, Inserm U1223, Paris, France
| |
Collapse
|
9
|
Peng X, Rao G, Li X, Tong N, Tian Y, Fu X. Preclinical models for Type 1 Diabetes Mellitus - A practical approach for research. Int J Med Sci 2023; 20:1644-1661. [PMID: 37859703 PMCID: PMC10583179 DOI: 10.7150/ijms.86566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Accepted: 09/18/2023] [Indexed: 10/21/2023] Open
Abstract
Numerous preclinical models have been developed to advance biomedical research in type 1 diabetes mellitus (T1DM). They are essential for improving our knowledge of T1DM development and progression, allowing researchers to identify potential therapeutic targets and evaluate the effectiveness of new medications. A deeper comprehension of these models themselves is critical not only to determine the optimal strategies for their utilization but also to fully unlock their potential applications in both basic and translational research. Here, we will comprehensively summarize and discuss the applications, advantages, and limitations of the commonly used animal models for human T1DM and also overview the up-to-date human tissue bioengineering models for the investigation of T1DM. By combining these models with a better understanding of the pathophysiology of T1DM, we can enhance our insights into disease initiation and development, ultimately leading to improved therapeutic responses and outcomes.
Collapse
Affiliation(s)
| | | | | | | | | | - Xianghui Fu
- Department of Endocrinology and Metabolism, Center for Diabetes Metabolism Research, Cancer Center West China Hospital, West China School of Medicine, Sichuan University, Chengdu, China
| |
Collapse
|
10
|
Abstract
Breakthroughs in cancer treatment with immunotherapeutics have provided long-term patient benefits for many different types of cancer. However, complete response is not achieved in many patients and tumor types, and the mechanisms underlying this lack of response are poorly understood. Despite this, numerous new targets, therapeutics, and drug combinations are being developed and tested in clinical trials. Preclinical models that recapitulate the complex human tumor microenvironment and the interplay between tumor and immune cells within the cancer-immunity cycle are needed to improve our understanding and screen new therapeutics for efficacy and safety/toxicity. Humanized mice, encompassing human tumors and human immune cells engrafted on immunodeficient mice, have been widely used for many years in immuno-oncology, with developments to improve both the humanization and the translational value central to the next generation of models. In this overview, we discuss recent advances in humanized models relevant to immuno-oncology drug discovery, the advantages and limitations of such models, the application of humanized models for efficacy and safety assessments of immunotherapeutics, and the potential opportunities. © 2023 Crown Bioscience. Current Protocols published by Wiley Periodicals LLC.
Collapse
Affiliation(s)
| | - Gerold Feuer
- Crown Bioscience Inc., San Diego, California, USA
| | | |
Collapse
|
11
|
Shu Y, Peng F, Zhao B, Liu C, Li Q, Li H, Wang Y, Jiang Y, Lu T, Wang Q, Sun J, Feng H, Lu Z, Liu X, Wang J, Qiu W. Transfer of patient's peripheral blood mononuclear cells (PBMCs) disrupts blood-brain barrier and induces anti-NMDAR encephalitis: a study of novel humanized PBMC mouse model. J Neuroinflammation 2023; 20:164. [PMID: 37443034 DOI: 10.1186/s12974-023-02844-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 06/29/2023] [Indexed: 07/15/2023] Open
Abstract
BACKGROUND Anti-N-methyl-D-aspartate receptor (NMDAR) encephalitis is a severe autoimmune neuropsychiatric disease. Brain access of anti-NMDAR autoantibody through the blood-brain barrier (BBB) is essential for pathogenesis. Most previous animal models limit the investigation of etiologies of BBB damage in patients. METHODS In this study, we established a novel humanized mouse model of anti-NMDAR encephalitis by intraperitoneal injection of patients' peripheral blood mononuclear cells (PBMCs) into BALB/c Rag2-/-Il2rg-/-SirpαNODFlk2-/- mice. RESULTS We found that engraftment of patients' PBMCs not only produced potent anti-GluN1 autoantibodies, but also disrupted BBB integrity to allow brain access of autoantibodies, resulting in a hyperactive locomotor phenotype, anxiety- and depressive-like behaviors, cognitive deficits, as well as functional changes in corresponding brain regions. Transcriptome analysis suggested an exaggerated immune response and impaired neurotransmission in the mouse model and highlighted Il-1β as a hub gene implicated in pathological changes. We further demonstrated that Il-1β was produced by endothelial cells and disrupted BBB by repressing tight junction proteins. Treatment with Anakinra, an Il-1 receptor antagonist, ameliorated BBB damage and neuropsychiatric behaviors. CONCLUSIONS Our study provided a novel and clinically more relevant humanized mouse model of anti-NMDAR encephalitis and revealed an intrinsic pathogenic property of the patient's lymphocytes.
Collapse
Affiliation(s)
- Yaqing Shu
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510630, China
| | - Fuhua Peng
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510630, China
| | - Bingchu Zhao
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences-Wuhan National Laboratory for Optoelectronics, Wuhan, 430071, China
| | - Chunxin Liu
- Department of Emergency, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Qihui Li
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510630, China
| | - Huilu Li
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510630, China
| | - Yuge Wang
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510630, China
| | - Yanjun Jiang
- Department of Anaesthesia and Intensive Care, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, Hong Kong SAR, China
- Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Tingting Lu
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510630, China
| | - Qin Wang
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Jian Sun
- Guangdong Laboratory for Lingnan Modern Agriculture, National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original BacteriaCollege of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Huiyu Feng
- Department of Neurology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Zhengqi Lu
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510630, China
| | - Xiaodong Liu
- Department of Anaesthesia and Intensive Care, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China.
- Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, Hong Kong SAR, China.
- Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China.
| | - Jie Wang
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences-Wuhan National Laboratory for Optoelectronics, Wuhan, 430071, China.
- Institute of Neuroscience and Brain Diseases; Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Wei Qiu
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510630, China.
| |
Collapse
|
12
|
Chen A, Neuwirth I, Herndler-Brandstetter D. Modeling the Tumor Microenvironment and Cancer Immunotherapy in Next-Generation Humanized Mice. Cancers (Basel) 2023; 15:2989. [PMID: 37296949 PMCID: PMC10251926 DOI: 10.3390/cancers15112989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 05/10/2023] [Accepted: 05/28/2023] [Indexed: 06/12/2023] Open
Abstract
Cancer immunotherapy has brought significant clinical benefits to numerous patients with malignant disease. However, only a fraction of patients experiences complete and durable responses to currently available immunotherapies. This highlights the need for more effective immunotherapies, combination treatments and predictive biomarkers. The molecular properties of a tumor, intratumor heterogeneity and the tumor immune microenvironment decisively shape tumor evolution, metastasis and therapy resistance and are therefore key targets for precision cancer medicine. Humanized mice that support the engraftment of patient-derived tumors and recapitulate the human tumor immune microenvironment of patients represent a promising preclinical model to address fundamental questions in precision immuno-oncology and cancer immunotherapy. In this review, we provide an overview of next-generation humanized mouse models suitable for the establishment and study of patient-derived tumors. Furthermore, we discuss the opportunities and challenges of modeling the tumor immune microenvironment and testing a variety of immunotherapeutic approaches using human immune system mouse models.
Collapse
Affiliation(s)
| | | | - Dietmar Herndler-Brandstetter
- Center for Cancer Research, Medical University of Vienna and Comprehensive Cancer Center, 1090 Vienna, Austria; (A.C.); (I.N.)
| |
Collapse
|
13
|
Zhao Y, Fang L, Guo P, Fang Y, Wu J. A MD Simulation Prediction for Regulation of N-Terminal Modification on Binding of CD47 to CD172a in a Force-Dependent Manner. Molecules 2023; 28:molecules28104224. [PMID: 37241964 DOI: 10.3390/molecules28104224] [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: 04/16/2023] [Revised: 05/12/2023] [Accepted: 05/18/2023] [Indexed: 05/28/2023] Open
Abstract
Cancer cells can evade immune surveillance through binding of its transmembrane receptor CD47 to CD172a on myeloid cells. CD47 is recognized as a promising immune checkpoint for cancer immunotherapy inhibiting macrophage phagocytosis. N-terminal post-translated modification (PTM) via glutaminyl cyclase is a landmark event in CD47 function maturation, but the molecular mechanism underlying the mechano-chemical regulation of the modification on CD47/CD172a remains unclear. Here, we performed so-called "ramp-clamp" steered molecular dynamics (SMD) simulations, and found that the N-terminal PTM enhanced interaction of CD172a with CD47 by inducing a dynamics-driven contraction of the binding pocket of the bound CD172a, an additional constraint on CYS15 on CD47 significantly improved the tensile strength of the complex with or without PTM, and a catch bond phenomenon would occur in complex dissociation under tensile force of 25 pN in a PTM-independent manner too. The residues GLN52 and SER66 on CD172a reinforced the H-bonding with their partners on CD47 in responding to PTM, while ARG69 on CD172 with its partner on CD47 might be crucial in the structural stability of the complex. This work might serve as molecular basis for the PTM-induced function improvement of CD47, should be helpful for deeply understanding CD47-relevant immune response and cancer development, and provides a novel insight in developing of new strategies of immunotherapy targeting this molecule interaction.
Collapse
Affiliation(s)
- Yang Zhao
- Institute of Biomechanics, School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| | - Liping Fang
- Institute of Biomechanics, School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| | - Pei Guo
- Institute of Biomechanics, School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| | - Ying Fang
- Institute of Biomechanics, School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| | - Jianhua Wu
- Institute of Biomechanics, School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| |
Collapse
|
14
|
Zhou Y, Xia J, Xu S, She T, Zhang Y, Sun Y, Wen M, Jiang T, Xiong Y, Lei J. Experimental mouse models for translational human cancer research. Front Immunol 2023; 14:1095388. [PMID: 36969176 PMCID: PMC10036357 DOI: 10.3389/fimmu.2023.1095388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 02/20/2023] [Indexed: 03/12/2023] Open
Abstract
The development and growth of tumors remains an important and ongoing threat to human life around the world. While advanced therapeutic strategies such as immune checkpoint therapy and CAR-T have achieved astonishing progress in the treatment of both solid and hematological malignancies, the malignant initiation and progression of cancer remains a controversial issue, and further research is urgently required. The experimental animal model not only has great advantages in simulating the occurrence, development, and malignant transformation mechanisms of tumors, but also can be used to evaluate the therapeutic effects of a diverse array of clinical interventions, gradually becoming an indispensable method for cancer research. In this paper, we have reviewed recent research progress in relation to mouse and rat models, focusing on spontaneous, induced, transgenic, and transplantable tumor models, to help guide the future study of malignant mechanisms and tumor prevention.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Tao Jiang
- *Correspondence: Jie Lei, ; Yanlu Xiong, ; Tao Jiang,
| | - Yanlu Xiong
- *Correspondence: Jie Lei, ; Yanlu Xiong, ; Tao Jiang,
| | - Jie Lei
- *Correspondence: Jie Lei, ; Yanlu Xiong, ; Tao Jiang,
| |
Collapse
|
15
|
Chuprin J, Buettner H, Seedhom MO, Greiner DL, Keck JG, Ishikawa F, Shultz LD, Brehm MA. Humanized mouse models for immuno-oncology research. Nat Rev Clin Oncol 2023; 20:192-206. [PMID: 36635480 PMCID: PMC10593256 DOI: 10.1038/s41571-022-00721-2] [Citation(s) in RCA: 109] [Impact Index Per Article: 54.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/12/2022] [Indexed: 01/14/2023]
Abstract
Immunotherapy has emerged as a promising treatment paradigm for many malignancies and is transforming the drug development landscape. Although immunotherapeutic agents have demonstrated clinical efficacy, they are associated with variable clinical responses, and substantial gaps remain in our understanding of their mechanisms of action and specific biomarkers of response. Currently, the number of preclinical models that faithfully recapitulate interactions between the human immune system and tumours and enable evaluation of human-specific immunotherapies in vivo is limited. Humanized mice, a term that refers to immunodeficient mice co-engrafted with human tumours and immune components, provide several advantages for immuno-oncology research. In this Review, we discuss the benefits and challenges of the currently available humanized mice, including specific interactions between engrafted human tumours and immune components, the development and survival of human innate immune populations in these mice, and approaches to study mice engrafted with matched patient tumours and immune cells. We highlight the latest advances in the generation of humanized mouse models, with the aim of providing a guide for their application to immuno-oncology studies with potential for clinical translation.
Collapse
Affiliation(s)
- Jane Chuprin
- Program in Molecular Medicine, The University of Massachusetts Chan Medical School, Worcester, MA, USA
- Department of Molecular, Cell and Cancer Biology, The University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Hannah Buettner
- Program in Molecular Medicine, The University of Massachusetts Chan Medical School, Worcester, MA, USA
- Department of Surgery, The University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Mina O Seedhom
- Program in Molecular Medicine, The University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Dale L Greiner
- Program in Molecular Medicine, The University of Massachusetts Chan Medical School, Worcester, MA, USA
| | | | | | | | - Michael A Brehm
- Program in Molecular Medicine, The University of Massachusetts Chan Medical School, Worcester, MA, USA.
| |
Collapse
|
16
|
Lanis JM, Lewis MS, Strassburger H, Larsen K, Bagby SM, Dominguez ATA, Marín-Jiménez JA, Pelanda R, Pitts TM, Lang J. Testing Cancer Immunotherapeutics in a Humanized Mouse Model Bearing Human Tumors. J Vis Exp 2022:10.3791/64606. [PMID: 36591990 PMCID: PMC11167650 DOI: 10.3791/64606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Reversing the immunosuppressive nature of the tumor microenvironment is critical for the successful treatment of cancers with immunotherapy drugs. Murine cancer models are extremely limited in their diversity and suffer from poor translation to the clinic. To serve as a more physiological preclinical model for immunotherapy studies, this protocol has been developed to evaluate the treatment of human tumors in a mouse reconstituted with a human immune system. This unique protocol demonstrates the development of human immune system (HIS, "humanized") mice, followed by implantation of a human tumor, either a cell-line derived xenograft (CDX) or a patient derived xenograft (PDX). HIS mice are generated by injecting CD34+ human hematopoietic stem cells isolated from umbilical cord blood into neonatal BRGS (BALB/c Rag2-/- IL2RγC-/- NODSIRPα) highly immunodeficient mice that are also capable of accepting a xenogeneic tumor. The importance of the kinetics and characteristics of the human immune system development and tumor implantation is emphasized. Finally, an in-depth evaluation of the tumor microenvironment using flow cytometry is described. In numerous studies using this protocol, it was found that the tumor microenvironment of individual tumors is recapitulated in HIS-PDX mice; "hot" tumors exhibit large immune infiltration while "cold" tumors do not. This model serves as a testing ground for combination immunotherapies for a wide range of human tumors and represents an important tool in the quest for personalized medicine.
Collapse
Affiliation(s)
- Jordi M Lanis
- Department of Immunology and Microbiology, School of Medicine, University of Colorado Denver Anschutz Medical Campus
| | - Matthew S Lewis
- Department of Immunology and Microbiology, School of Medicine, University of Colorado Denver Anschutz Medical Campus
| | - Hannah Strassburger
- Department of Immunology and Microbiology, School of Medicine, University of Colorado Denver Anschutz Medical Campus
| | - Kristina Larsen
- Department of Immunology and Microbiology, School of Medicine, University of Colorado Denver Anschutz Medical Campus
| | - Stacey M Bagby
- Division of Oncology, School of Medicine, University of Colorado Denver Anschutz Medical Campus
| | - Adrian T A Dominguez
- Division of Oncology, School of Medicine, University of Colorado Denver Anschutz Medical Campus
| | - Juan A Marín-Jiménez
- Department of Medical Oncology, Catalan Institute of Oncology (ICO-L'Hospitalet)
| | - Roberta Pelanda
- Department of Immunology and Microbiology, School of Medicine, University of Colorado Denver Anschutz Medical Campus
| | - Todd M Pitts
- Division of Oncology, School of Medicine, University of Colorado Denver Anschutz Medical Campus
| | - Julie Lang
- Department of Immunology and Microbiology, School of Medicine, University of Colorado Denver Anschutz Medical Campus;
| |
Collapse
|
17
|
Wang X, Wu C, Wei H. Humanized Germ-Free Mice for Investigating the Intervention Effect of Commensal Microbiome on Cancer Immunotherapy. Antioxid Redox Signal 2022; 37:1291-1302. [PMID: 35403435 DOI: 10.1089/ars.2022.0039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Significance: A growing body of evidence has demonstrated that the commensal microbiome is deeply involved in the host immune response, accounting for significantly divergent clinical outcomes among cancer patients receiving immunotherapy. Therefore, precise screening and evaluating of functional bacterial strains as novel targets for cancer immunotherapy have attracted great enthusiasm from both academia and industry, which calls for the construction and application of advanced animal models to support translational research in this field. Recent Advances: Significant progress has been made to elucidate the intervention effect of commensal microbiome on immunotherapy based on animal experiments. Especially, correlation between gut microbiota and host response to immunotherapy has been continuously discovered in a variety of cancer types, laying the foundation for causality establishment and mechanism research. Critical Issues: In oncology research, it is particularly not uncommon to see that a promising preclinical result fails to translate into clinical success. The use of conventional murine models in immunotherapy-associated microbiome research is very likely to bring discredit on the preclinical findings. We emphasize the value of germ-free (GF) mice and humanized mice as advanced models in this field. Future Directions: Integrating rederivation and humanization to generate humanized GF mice as preclinical models would make it possible to clarify the role of specific bacterial strains in immunotherapy as well as obtain preclinical findings that are more predictive for humans, leading to novel microbiome-based strategies for cancer immunotherapy. Antioxid. Redox Signal. 37, 1291-1302.
Collapse
Affiliation(s)
- Xinning Wang
- Precision Medicine Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Chengwei Wu
- Precision Medicine Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hong Wei
- Precision Medicine Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
18
|
Chen J, Liao S, Xiao Z, Pan Q, Wang X, Shen K, Wang S, Yang L, Guo F, Liu HF, Pan Q. The development and improvement of immunodeficient mice and humanized immune system mouse models. Front Immunol 2022; 13:1007579. [PMID: 36341323 PMCID: PMC9626807 DOI: 10.3389/fimmu.2022.1007579] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 10/07/2022] [Indexed: 12/02/2022] Open
Abstract
Animal models play an indispensable role in the study of human diseases. However, animal models of different diseases do not fully mimic the complex internal environment of humans. Immunodeficient mice are deficient in certain genes and do not express these or show reduced expression in some of their cells, facilitating the establishment of humanized mice and simulation of the human environment in vivo. Here, we summarize the developments in immunodeficient mice, from the initial nude mice lacking T lymphocytes to NOD/SCID rgnull mice lacking T, B, and NK cell populations. We describe existing humanized immune system mouse models based on immunodeficient mice in which human cells or tissues have been transplanted to establish a human immune system, including humanized-peripheral blood mononuclear cells (Hu-PBMCs), humanized hematopoietic stem cells (Hu-HSCs), and humanized bone marrow, liver, thymus (Hu-BLT) mouse models. The different methods for their development involve varying levels of complexity and humanization. Humanized mice are widely used in the study of various diseases to provide a transitional stage for clinical research. However, several challenges persist, including improving the efficiency of reconstructing the human B cell immune response, extending lifespan, improving the survival rate of mice to extend the observation period, and improving the development of standardized commercialized models and as well as their use. Overall, there are many opportunities and challenges in the development of humanized immune system mouse models which can provide novel strategies for understanding the mechanisms and treatments of human disease.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Qingjun Pan
- *Correspondence: Hua-feng Liu, ; Qingjun Pan,
| |
Collapse
|
19
|
Nath PR, Pal-Nath D, Kaur S, Gangaplara A, Meyer TJ, Cam MC, Roberts DD. Loss of CD47 alters CD8+ T cell activation in vitro and immunodynamics in mice. Oncoimmunology 2022; 11:2111909. [PMID: 36105746 PMCID: PMC9467551 DOI: 10.1080/2162402x.2022.2111909] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 08/03/2022] [Accepted: 08/07/2022] [Indexed: 12/03/2022] Open
Abstract
CD47 has established roles in the immune system for regulating macrophage phagocytosis and lymphocyte activation, with growing evidence of its cell-intrinsic regulatory roles in natural killer and CD8+ T cells. CD47 limits antigen-dependent cytotoxic activities of human and murine CD8+ T cells, but its role in T cell activation kinetics remains unclear. Using in vitro and in vivo models, we show here that CD47 differentially regulates CD8+ T cell responses to short- versus long-term activation. Although CD47 was not required for T cell development in mice and early activation in vitro, short-term stimuli elevated pathogen-reactive gene expression and enhanced proliferation and the effector phenotypes of Cd47-deficient relative to Cd47-sufficient CD8+ T cells. In contrast, persistent TCR stimulation limited the effector phenotypes of Cd47 -/- CD8+ T cells and enhanced their apoptosis signature. CD8+ T cell expansion and activation in vivo induced by acute lymphocytic choriomeningitis virus (LCMV) infection did not differ in the absence of CD47. However, the frequency and effector phenotypes of Cd47-/- CD8+ T cells were constrained in chronic LCMV-infected as well as in mice bearing B16 melanoma tumors. Therefore, CD47 regulates CD8+ T cell activation, proliferation, and fitness in a context-dependent manner.
Collapse
Affiliation(s)
- Pulak R. Nath
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- Clinical and Translational Immunology Unit, Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Dipasmita Pal-Nath
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Sukhbir Kaur
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Arunkumar Gangaplara
- Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Thomas J. Meyer
- CCR Collaborative Bioinformatics Resource, Office of Science and Technology Resources, National Cancer Institute, Bethesda, MD, USA
| | - Margaret C Cam
- CCR Collaborative Bioinformatics Resource, Office of Science and Technology Resources, National Cancer Institute, Bethesda, MD, USA
| | - David D. Roberts
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
20
|
Chan C, Lustig M, Baumann N, Valerius T, van Tetering G, Leusen JHW. Targeting Myeloid Checkpoint Molecules in Combination With Antibody Therapy: A Novel Anti-Cancer Strategy With IgA Antibodies? Front Immunol 2022; 13:932155. [PMID: 35865547 PMCID: PMC9295600 DOI: 10.3389/fimmu.2022.932155] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 06/07/2022] [Indexed: 11/13/2022] Open
Abstract
Immunotherapy with therapeutic antibodies has shown a lack of durable responses in some patients due to resistance mechanisms. Checkpoint molecules expressed by tumor cells have a deleterious impact on clinical responses to therapeutic antibodies. Myeloid checkpoints, which negatively regulate macrophage and neutrophil anti-tumor responses, are a novel type of checkpoint molecule. Myeloid checkpoint inhibition is currently being studied in combination with IgG-based immunotherapy. In contrast, the combination with IgA-based treatment has received minimal attention. IgA antibodies have been demonstrated to more effectively attract and activate neutrophils than their IgG counterparts. Therefore, myeloid checkpoint inhibition could be an interesting addition to IgA treatment and has the potential to significantly enhance IgA therapy.
Collapse
Affiliation(s)
- Chilam Chan
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Marta Lustig
- Division of Stem Cell Transplantation and Immunotherapy, Department of Medicine II, Christian Albrechts University Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Niklas Baumann
- Division of Stem Cell Transplantation and Immunotherapy, Department of Medicine II, Christian Albrechts University Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Thomas Valerius
- Division of Stem Cell Transplantation and Immunotherapy, Department of Medicine II, Christian Albrechts University Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Geert van Tetering
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Jeanette H. W. Leusen
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands
- *Correspondence: Jeanette H. W. Leusen,
| |
Collapse
|
21
|
Aparici Herraiz I, Caires HR, Castillo-Fernández Ó, Sima N, Méndez-Mora L, Risueño RM, Sattabongkot J, Roobsoong W, Hernández-Machado A, Fernandez-Becerra C, Barrias CC, del Portillo HA. Advancing Key Gaps in the Knowledge of Plasmodium vivax Cryptic Infections Using Humanized Mouse Models and Organs-on-Chips. Front Cell Infect Microbiol 2022; 12:920204. [PMID: 35873153 PMCID: PMC9302440 DOI: 10.3389/fcimb.2022.920204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 06/06/2022] [Indexed: 11/13/2022] Open
Abstract
Plasmodium vivax is the most widely distributed human malaria parasite representing 36.3% of disease burden in the South-East Asia region and the most predominant species in the region of the Americas. Recent estimates indicate that 3.3 billion of people are under risk of infection with circa 7 million clinical cases reported each year. This burden is certainly underestimated as the vast majority of chronic infections are asymptomatic. For centuries, it has been widely accepted that the only source of cryptic parasites is the liver dormant stages known as hypnozoites. However, recent evidence indicates that niches outside the liver, in particular in the spleen and the bone marrow, can represent a major source of cryptic chronic erythrocytic infections. The origin of such chronic infections is highly controversial as many key knowledge gaps remain unanswered. Yet, as parasites in these niches seem to be sheltered from immune response and antimalarial drugs, research on this area should be reinforced if elimination of malaria is to be achieved. Due to ethical and technical considerations, working with the liver, bone marrow and spleen from natural infections is very difficult. Recent advances in the development of humanized mouse models and organs-on-a-chip models, offer novel technological frontiers to study human diseases, vaccine validation and drug discovery. Here, we review current data of these frontier technologies in malaria, highlighting major challenges ahead to study P. vivax cryptic niches, which perpetuate transmission and burden.
Collapse
Affiliation(s)
- Iris Aparici Herraiz
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
- Institut d’Investigació en Ciències de la Salut Germans Trias i Pujol, Badalona, Spain
| | - Hugo R. Caires
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Óscar Castillo-Fernández
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
- Institut d’Investigació en Ciències de la Salut Germans Trias i Pujol, Badalona, Spain
- Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona, Barcelona, Spain
| | - Núria Sima
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
- Institut d’Investigació en Ciències de la Salut Germans Trias i Pujol, Badalona, Spain
| | - Lourdes Méndez-Mora
- Department of Condensed Matter Physics, University of Barcelona (UB), Barcelona, Spain
| | - Ruth M. Risueño
- Josep Carreras Leukaemia Research Institute (IJC), Barcelona, Spain
| | - Jetsumon Sattabongkot
- Mahidol Vivax Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Wanlapa Roobsoong
- Mahidol Vivax Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Aurora Hernández-Machado
- Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona, Barcelona, Spain
- Department of Condensed Matter Physics, University of Barcelona (UB), Barcelona, Spain
- Centre de Recerca Matemàtica (CRM), Barcelona, Spain
| | - Carmen Fernandez-Becerra
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
- Institut d’Investigació en Ciències de la Salut Germans Trias i Pujol, Badalona, Spain
| | - Cristina C. Barrias
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- INEB – Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
- ICBAS – Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Hernando A. del Portillo
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
- Institut d’Investigació en Ciències de la Salut Germans Trias i Pujol, Badalona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
- *Correspondence: Hernando A. del Portillo,
| |
Collapse
|
22
|
van Duijn A, Van der Burg SH, Scheeren FA. CD47/SIRPα axis: bridging innate and adaptive immunity. J Immunother Cancer 2022; 10:jitc-2022-004589. [PMID: 35831032 PMCID: PMC9280883 DOI: 10.1136/jitc-2022-004589] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/09/2022] [Indexed: 11/29/2022] Open
Abstract
Myeloid immune cells are frequently present in the tumor environment, and although they can positively contribute to tumor control they often negatively impact anticancer immune responses. One way of inhibiting the positive contributions of myeloid cells is by signaling through the cluster of differentiation 47 (CD47)/signal regulatory protein alpha (SIRPα) axis. The SIRPα receptor is expressed on myeloid cells and is an inhibitory immune receptor that, upon binding to CD47 protein, delivers a ‘don’t eat me’ signal. As CD47 is often overexpressed on cancer cells, treatments targeting CD47/SIRPα have been under active investigation and are currently being tested in clinical settings. Interestingly, the CD47/SIRPα axis is also involved in T cell-mediated antitumor responses. In this perspective we provide an overview of recent studies showing how therapeutic blockade of the CD47/SIRPα axis improves the adaptive immune response. Furthermore, we discuss the interconnection between the myeloid CD47/SIRPα axis and adaptive T cell responses as well as the potential therapeutic role of the CD47/SIRPα axis in tumors with acquired resistance to the classic immunotherapy through major histocompatibility complex downregulation. Altogether this review provides a profound insight for the optimal exploitation of CD47/SIRPα immune checkpoint therapy.
Collapse
Affiliation(s)
- Anneloes van Duijn
- Department of Medical Oncology, Leiden University Medical Center, Leiden, The Netherlands.,Department of Dermatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Sjoerd H Van der Burg
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| | - Ferenc A Scheeren
- Department of Dermatology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
23
|
Zhang J, Xu Y, Zhang Y, Bossila EA, Shi M, Zhao Y. Bioinformatic analysis as a first step to predict the compatibility of hematopoiesis and immune system genes between humans and pigs. Xenotransplantation 2022; 29:e12764. [PMID: 35695327 DOI: 10.1111/xen.12764] [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: 02/15/2022] [Revised: 05/17/2022] [Accepted: 05/20/2022] [Indexed: 11/29/2022]
Abstract
The shortage of allogeneic donor organs leaves its supply far short of clinical need. There are great expectations on xenotransplantation, especially with pigs' organs. With the genetic modification of donor pigs, the rejection and cross-species transmission issues have now been widely addressed. However, research on the compatibility of genes between humans and pigs was limited. We performed a systematic screening analysis of predicted incompatible genes between humans and pigs, judged by low protein sequence similarities or different predicted protein domain compositions. By combining with gene set enrichment analysis, we screened out several key genes of hematopoiesis and the immune system with possible incompatibilities, which might be important for establishing chimera and xenotransplantation between humans and pigs. There were seven chemokine genes, including CCL1, CCL5, CCL24, CCL25, CCL28, CXCL12, and CXCL16, that exhibited limited similarity between humans and pigs (similarity < 0.8). Among hematopoiesis process-related genes, 15 genes of adhesion molecules, Notch ligands, and cytokine receptors exhibited differences between humans and pigs. In complement and coagulation cascades, 19 genes showed low similarity and 77 genes had different domain compositions between humans and pigs. Our study provides a good reference for further genetic modification of pigs, which might be beneficial for xenotransplantation.
Collapse
Affiliation(s)
- Jiayu Zhang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Yanan Xu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Yingzi Zhang
- Biological and Environmental Sciences and Engineering Division, King Abdullah University of Science and Technology, Thuwal, Saudi Arabia
| | - Elhusseny A Bossila
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China.,Biotechnology Department, Faculty of Agriculture Al-Azhar University, Cairo, Egypt
| | - Mingpu Shi
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Yong Zhao
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
24
|
Yiu YY, Hansen PS, Torrez Dulgeroff LB, Blacker G, Myers L, Galloway S, Gars E, Colace O, Mansfield P, Hasenkrug KJ, Weissman IL, Tal MC. CD47 Blockade Leads to Chemokine-Dependent Monocyte Infiltration and Loss of B Cells from the Splenic Marginal Zone. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:1371-1377. [PMID: 35236754 PMCID: PMC9012117 DOI: 10.4049/jimmunol.2100352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 01/11/2022] [Indexed: 11/19/2022]
Abstract
CD47 is an important innate immune checkpoint through its interaction with its inhibitory receptor on macrophages, signal-regulatory protein α (SIRPα). Therapeutic blockade of CD47-SIRPα interactions is a promising immuno-oncology treatment that promotes clearance of cancer cells. However, CD47-SIRPα interactions also maintain homeostatic lymphocyte levels. In this study, we report that the mouse splenic marginal zone B cell population is dependent on intact CD47-SIRPα interactions and blockade of CD47 leads to the loss of these cells. This depletion is accompanied by elevated levels of monocyte-recruiting chemokines CCL2 and CCL7 and infiltration of CCR2+Ly6Chi monocytes into the mouse spleen. In the absence of CCR2 signaling, there is no infiltration and reduced marginal zone B cell depletion. These data suggest that CD47 blockade leads to clearance of splenic marginal zone B cells.
Collapse
Affiliation(s)
- Ying Ying Yiu
- Institute for Stem Cell Biology and Regenerative Medicine and the Ludwig Cancer Center, Stanford University School of Medicine, Stanford, CA
- Immunology Program, Stanford University, Stanford, CA
| | - Paige S Hansen
- Institute for Stem Cell Biology and Regenerative Medicine and the Ludwig Cancer Center, Stanford University School of Medicine, Stanford, CA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA
| | - Laughing Bear Torrez Dulgeroff
- Institute for Stem Cell Biology and Regenerative Medicine and the Ludwig Cancer Center, Stanford University School of Medicine, Stanford, CA
| | - Grace Blacker
- Institute for Stem Cell Biology and Regenerative Medicine and the Ludwig Cancer Center, Stanford University School of Medicine, Stanford, CA
| | - Lara Myers
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT
| | - Sarah Galloway
- Institute for Stem Cell Biology and Regenerative Medicine and the Ludwig Cancer Center, Stanford University School of Medicine, Stanford, CA
| | - Eric Gars
- Department of Pathology, Stanford University School of Medicine, Stanford, CA
| | - Olivia Colace
- Institute for Stem Cell Biology and Regenerative Medicine and the Ludwig Cancer Center, Stanford University School of Medicine, Stanford, CA
| | - Paul Mansfield
- Institute for Stem Cell Biology and Regenerative Medicine and the Ludwig Cancer Center, Stanford University School of Medicine, Stanford, CA
| | - Kim J Hasenkrug
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT
| | - Irving L Weissman
- Institute for Stem Cell Biology and Regenerative Medicine and the Ludwig Cancer Center, Stanford University School of Medicine, Stanford, CA;
- Department of Pathology, Stanford University School of Medicine, Stanford, CA
- Ludwig Center for Cancer Stem Cell Research and Medicine, Stanford University School of Medicine, Stanford, CA; and
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA
| | - Michal Caspi Tal
- Institute for Stem Cell Biology and Regenerative Medicine and the Ludwig Cancer Center, Stanford University School of Medicine, Stanford, CA;
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA
| |
Collapse
|
25
|
Khosravi-Maharlooei M, Madley R, Borsotti C, Ferreira LMR, Sharp RC, Brehm MA, Greiner DL, Parent AV, Anderson MS, Sykes M, Creusot RJ. Modeling human T1D-associated autoimmune processes. Mol Metab 2022; 56:101417. [PMID: 34902607 PMCID: PMC8739876 DOI: 10.1016/j.molmet.2021.101417] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 11/19/2021] [Accepted: 12/07/2021] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Type 1 diabetes (T1D) is an autoimmune disease characterized by impaired immune tolerance to β-cell antigens and progressive destruction of insulin-producing β-cells. Animal models have provided valuable insights for understanding the etiology and pathogenesis of this disease, but they fall short of reflecting the extensive heterogeneity of the disease in humans, which is contributed by various combinations of risk gene alleles and unique environmental factors. Collectively, these factors have been used to define subgroups of patients, termed endotypes, with distinct predominating disease characteristics. SCOPE OF REVIEW Here, we review the gaps filled by these models in understanding the intricate involvement and regulation of the immune system in human T1D pathogenesis. We describe the various models developed so far and the scientific questions that have been addressed using them. Finally, we discuss the limitations of these models, primarily ascribed to hosting a human immune system (HIS) in a xenogeneic recipient, and what remains to be done to improve their physiological relevance. MAJOR CONCLUSIONS To understand the role of genetic and environmental factors or evaluate immune-modifying therapies in humans, it is critical to develop and apply models in which human cells can be manipulated and their functions studied under conditions that recapitulate as closely as possible the physiological conditions of the human body. While microphysiological systems and living tissue slices provide some of these conditions, HIS mice enable more extensive analyses using in vivo systems.
Collapse
Affiliation(s)
- Mohsen Khosravi-Maharlooei
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Rachel Madley
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Chiara Borsotti
- Department of Health Sciences, Histology laboratory, Università del Piemonte Orientale, Novara, Italy
| | - Leonardo M R Ferreira
- Departments of Microbiology & Immunology, and Regenerative Medicine & Cell Biology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
| | - Robert C Sharp
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida College of Medicine, Gainesville, FL, USA
| | - Michael A Brehm
- Program in Molecular Medicine, Diabetes Center of Excellence, University of Massachusetts Medical School, Worcester, MA, USA
| | - Dale L Greiner
- Program in Molecular Medicine, Diabetes Center of Excellence, University of Massachusetts Medical School, Worcester, MA, USA
| | - Audrey V Parent
- Diabetes Center, University of California San Francisco, San Francisco, CA, USA
| | - Mark S Anderson
- Diabetes Center, University of California San Francisco, San Francisco, CA, USA
| | - Megan Sykes
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Remi J Creusot
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA.
| |
Collapse
|
26
|
Labarthe L, Gelé T, Gouget H, Benzemrane MS, Le Calvez P, Legrand N, Lambotte O, Le Grand R, Bourgeois C, Barrail-Tran A. Pharmacokinetics and tissue distribution of tenofovir, emtricitabine and dolutegravir in mice. J Antimicrob Chemother 2022; 77:1094-1101. [PMID: 35022753 DOI: 10.1093/jac/dkab501] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 12/20/2021] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Studies of antiretroviral drug (ARV) tissue distribution in preclinical models, such as mice, are key to understanding viral persistence. OBJECTIVES To determine the plasma and tissue pharmacokinetics and tissue distributions of tenofovir, emtricitabine and dolutegravir in mice. METHODS ARVs were simultaneously administered to two different strains, and their levels in plasma and tissue samples were determined by a validated LC-MS/MS method. A non-compartmental analysis was performed to estimate the main pharmacokinetic parameters. A tissue penetration factor (TPF) was calculated as the ratio of the concentration in the tissue concerned to that in plasma. RESULTS ARV plasma pharmacokinetic parameters in both strains were similar to those estimated in the clinical context. Tissue concentrations were highest in the digestive tract, followed by the liver and kidneys, lymphatic system, pancreas, adipose tissue and lungs. Tissue concentrations were lowest in the brain. Triple therapy could not be considered effective in any of the tissues considered. The TPF values obtained showed that tenofovir diffused widely, especially in the digestive tract, liver and kidneys. Emtricitabine had a TPF above 100% in two-thirds of the tissues. Dolutegravir was poorly distributed to all tissues. CONCLUSIONS Drug specificity was observed, with higher levels of exposure to tenofovir than to emtricitabine or dolutegravir. Tissue specificity was also observed, with strong penetration of the digestive tract and weak penetration of the brain. These data have important implications for future preclinical and clinical studies for developing new HIV therapies with the goal of an HIV cure.
Collapse
Affiliation(s)
- Laura Labarthe
- Université Paris-Saclay, Inserm, CEA, Immunologie des maladies virales, auto-immunes, hématologiques et bactériennes, 92265, Fontenay-aux-Roses, France.,genOway Paris, 92265, Fontenay-aux-Roses, France
| | - Thibaut Gelé
- Université Paris-Saclay, AP-HP, Hôpital Bicêtre, UMR1184, Inserm, CEA, Le Kremlin-Bicêtre, France
| | - Hélène Gouget
- Université Paris-Saclay, Inserm, CEA, Immunologie des maladies virales, auto-immunes, hématologiques et bactériennes, 92265, Fontenay-aux-Roses, France
| | - Mariam-Sarah Benzemrane
- Université Paris-Saclay, Inserm, CEA, Immunologie des maladies virales, auto-immunes, hématologiques et bactériennes, 92265, Fontenay-aux-Roses, France
| | - Pauline Le Calvez
- Université Paris-Saclay, Inserm, CEA, Immunologie des maladies virales, auto-immunes, hématologiques et bactériennes, 92265, Fontenay-aux-Roses, France
| | | | - Olivier Lambotte
- Université Paris-Saclay, AP-HP, Hôpital Bicêtre, UMR1184, Inserm, CEA, Le Kremlin-Bicêtre, France
| | - Roger Le Grand
- Université Paris-Saclay, Inserm, CEA, Immunologie des maladies virales, auto-immunes, hématologiques et bactériennes, 92265, Fontenay-aux-Roses, France
| | - Christine Bourgeois
- Université Paris-Saclay, Inserm, CEA, Immunologie des maladies virales, auto-immunes, hématologiques et bactériennes, 92265, Fontenay-aux-Roses, France
| | - Aurélie Barrail-Tran
- Université Paris-Saclay, AP-HP, Hôpital Bicêtre, UMR1184, Inserm, CEA, Le Kremlin-Bicêtre, France
| |
Collapse
|
27
|
Ducret A, Ackaert C, Bessa J, Bunce C, Hickling T, Jawa V, Kroenke MA, Lamberth K, Manin A, Penny HL, Smith N, Terszowski G, Tourdot S, Spindeldreher S. Assay format diversity in pre-clinical immunogenicity risk assessment: Toward a possible harmonization of antigenicity assays. MAbs 2021; 14:1993522. [PMID: 34923896 PMCID: PMC8726688 DOI: 10.1080/19420862.2021.1993522] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
A major impediment to successful use of therapeutic protein drugs is their ability to induce anti-drug antibodies (ADA) that can alter treatment efficacy and safety in a significant number of patients. To this aim, in silico, in vitro, and in vivo tools have been developed to assess sequence and other liabilities contributing to ADA development at different stages of the immune response. However, variability exists between similar assays developed by different investigators due to the complexity of assays, a degree of uncertainty about the underlying science, and their intended use. The impact of protocol variations on the outcome of the assays, i.e., on the immunogenicity risk assigned to a given drug candidate, cannot always be precisely assessed. Here, the Non-Clinical Immunogenicity Risk Assessment working group of the European Immunogenicity Platform (EIP) reviews currently used assays and protocols and discusses feasibility and next steps toward harmonization and standardization.
Collapse
Affiliation(s)
- Axel Ducret
- Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center, Basel, Switzerland
| | - Chloé Ackaert
- ImmunXperts SA (A Nexelis Group Company), Gosselies, Belgium
| | - Juliana Bessa
- Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center, Basel, Switzerland
| | | | - Timothy Hickling
- Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center, Basel, Switzerland
| | - Vibha Jawa
- Biotherapeutics and Bioanalysis Non-Clinical Development, Bristol Myers Squibb, Princeton, NJ, USA
| | - Mark A Kroenke
- Clinical Immunology-Translational Medicine, Amgen Inc, Thousand Oaks, CA, USA
| | - Kasper Lamberth
- Analysis & Characterisation, Global Research Technologies, Novo Nordisk A/S, Måløv, Denmark
| | - Anaïs Manin
- Abzena, Babraham Research Campus, Cambridge, UK
| | - Hweixian L Penny
- Clinical Immunology-Translational Medicine, Amgen Inc, Thousand Oaks, CA, USA
| | - Noel Smith
- Lonza Biologics, Chesterford Research Park, Saffron Walden, UK
| | - Grzegorz Terszowski
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Basel, Switzerland
| | | | | |
Collapse
|
28
|
Guo M, Xia C, Wu Y, Zhou N, Chen Z, Li W. Research Progress on Cell Membrane-Coated Biomimetic Delivery Systems. Front Bioeng Biotechnol 2021; 9:772522. [PMID: 34869288 PMCID: PMC8636778 DOI: 10.3389/fbioe.2021.772522] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 10/29/2021] [Indexed: 01/12/2023] Open
Abstract
Cell membrane-coated biomimetic nanoplatforms have many inherent properties, such as bio-interfacing abilities, self-identification, and signal transduction, which enable the biomimetic delivery system to escape immune clearance and opsonization. This can also maximize the drug delivery efficiency of synthetic nanoparticles (NPs) and functional cell membranes. As a new type of delivery system, cell membrane-coated biomimetic delivery systems have broadened the prospects for biomedical applications. In this review, we summarize research progress on cell membrane biomimetic technology from three aspects, including sources of membrane, modifications, and applications, then analyze their limitations and propose future research directions.
Collapse
Affiliation(s)
- Mengyu Guo
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China.,Engineering Center of State Ministry of Education for Standardization of Chinese Medicine Processing, Nanjing University of Chinese Medicine, Nanjing, China
| | - Chenjie Xia
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China.,Engineering Center of State Ministry of Education for Standardization of Chinese Medicine Processing, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yu Wu
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China.,Engineering Center of State Ministry of Education for Standardization of Chinese Medicine Processing, Nanjing University of Chinese Medicine, Nanjing, China
| | - Nong Zhou
- The Chongqing Engineering Laboratory for Green Cultivation and Deep Processing of Three Gorges Reservoir Area's Medicinal Herbs, College of Food and Biology Engineering, Chongqing Three Gorges University, Chongqing, China
| | - Zhipeng Chen
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China.,Engineering Center of State Ministry of Education for Standardization of Chinese Medicine Processing, Nanjing University of Chinese Medicine, Nanjing, China
| | - Weidong Li
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China.,Engineering Center of State Ministry of Education for Standardization of Chinese Medicine Processing, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
29
|
Mesenchymal stromal cell apoptosis is required for their therapeutic function. Nat Commun 2021; 12:6495. [PMID: 34764248 PMCID: PMC8586224 DOI: 10.1038/s41467-021-26834-3] [Citation(s) in RCA: 105] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 10/21/2021] [Indexed: 12/21/2022] Open
Abstract
Multipotent mesenchymal stromal cells (MSCs) ameliorate a wide range of diseases in preclinical models, but the lack of clarity around their mechanisms of action has impeded their clinical utility. The therapeutic effects of MSCs are often attributed to bioactive molecules secreted by viable MSCs. However, we found that MSCs underwent apoptosis in the lung after intravenous administration, even in the absence of host cytotoxic or alloreactive cells. Deletion of the apoptotic effectors BAK and BAX prevented MSC death and attenuated their immunosuppressive effects in disease models used to define MSC potency. Mechanistically, apoptosis of MSCs and their efferocytosis induced changes in metabolic and inflammatory pathways in alveolar macrophages to effect immunosuppression and reduce disease severity. Our data reveal a mode of action whereby the host response to dying MSCs is key to their therapeutic effects; findings that have broad implications for the effective translation of cell-based therapies. Mesenchymal stromal cells (MSCs) demonstrate therapeutic benefits in multiple diseases, but the mechanisms remain unclear as infused MSCs do not persist in the body. Here, the authors show that MSC apoptosis is an important mechanistic element, as MSCs rendered genetically incapable of apoptosis lose their ability to ameliorate disease.
Collapse
|
30
|
Sharp RC, Brown ME, Shapiro MR, Posgai AL, Brusko TM. The Immunoregulatory Role of the Signal Regulatory Protein Family and CD47 Signaling Pathway in Type 1 Diabetes. Front Immunol 2021; 12:739048. [PMID: 34603322 PMCID: PMC8481641 DOI: 10.3389/fimmu.2021.739048] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 08/31/2021] [Indexed: 12/11/2022] Open
Abstract
Background The pathogenesis of type 1 diabetes (T1D) involves complex genetic susceptibility that impacts pathways regulating host immunity and the target of autoimmune attack, insulin-producing pancreatic β-cells. Interactions between risk variants and environmental factors result in significant heterogeneity in clinical presentation among those who develop T1D. Although genetic risk is dominated by the human leukocyte antigen (HLA) class II and insulin (INS) gene loci, nearly 150 additional risk variants are significantly associated with the disease, including polymorphisms in immune checkpoint molecules, such as SIRPG. Scope of Review In this review, we summarize the literature related to the T1D-associated risk variants in SIRPG, which include a protein-coding variant (rs6043409, G>A; A263V) and an intronic polymorphism (rs2281808, C>T), and their potential impacts on the immunoregulatory signal regulatory protein (SIRP) family:CD47 signaling axis. We discuss how dysregulated expression or function of SIRPs and CD47 in antigen-presenting cells (APCs), T cells, natural killer (NK) cells, and pancreatic β-cells could potentially promote T1D development. Major Conclusions We propose a hypothesis, supported by emerging genetic and functional immune studies, which states a loss of proper SIRP:CD47 signaling may result in increased lymphocyte activation and cytotoxicity and enhanced β-cell destruction. Thus, we present several novel therapeutic strategies for modulation of SIRPs and CD47 to intervene in T1D.
Collapse
MESH Headings
- Animals
- Antigen-Presenting Cells/immunology
- Antigen-Presenting Cells/metabolism
- Antigens, Differentiation/genetics
- Antigens, Differentiation/metabolism
- CD47 Antigen/metabolism
- Diabetes Mellitus, Type 1/genetics
- Diabetes Mellitus, Type 1/immunology
- Diabetes Mellitus, Type 1/metabolism
- Diabetes Mellitus, Type 1/therapy
- Genetic Association Studies
- Humans
- Immunotherapy
- Insulin-Secreting Cells/immunology
- Insulin-Secreting Cells/metabolism
- Killer Cells, Natural/immunology
- Killer Cells, Natural/metabolism
- Polymorphism, Genetic
- Receptors, Cell Surface/metabolism
- Receptors, Immunologic/genetics
- Receptors, Immunologic/metabolism
- Signal Transduction
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
Collapse
Affiliation(s)
- Robert C. Sharp
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Matthew E. Brown
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Melanie R. Shapiro
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Amanda L. Posgai
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Todd M. Brusko
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL, United States
- Department of Pediatrics, College of Medicine, Diabetes Institute, University of Florida, Gainesville, FL, United States
| |
Collapse
|
31
|
Winterling K, Martin WD, De Groot AS, Daufenbach J, Kistner S, Schüttrumpf J. Development of a novel fully functional coagulation factor VIII with reduced immunogenicity utilizing an in silico prediction and deimmunization approach. J Thromb Haemost 2021; 19:2161-2170. [PMID: 34060724 PMCID: PMC8456792 DOI: 10.1111/jth.15413] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 03/17/2021] [Accepted: 04/19/2021] [Indexed: 11/26/2022]
Abstract
BACKGROUND Up to 30% of hemophilia A patients develop inhibitory antibodies against the infused factor VIII (FVIII). The development of a deimmunized FVIII is an unmet high medical need. Although improved recombinant FVIII (rFVIII) products evolved within the last years, the immunogenicity has not been solved. A deimmunized FVIII could reduce the probability of inhibitor development, providing safer therapy. OBJECTIVE To develop a deimmunized FVIII molecule by modifying major histocompatibility complex (MHC) class II presentation, leading to a functional but less immunogenic molecule. METHODS We performed (1) in silico prediction of potentially immunogenic T cell epitopes and their modification by amino acid substitutions in the FVIII sequence, (2) evaluation of functional and structural similarity of the modified rFVIII to unmodified FVIII and registered products, and (3) confirmation of the reduced immunogenicity by in vitro testing. RESULTS A partially deimmunized fully functional FVIII molecule incorporating 19 amino acid substitutions was generated. The substitutions led to a reduction of the immunogenicity score, indicating a reduced immunogenicity based on in silico calculations. This was confirmed in an in vitro dendritic cell (DC)--T cell assay. Using this assay, cells from healthy donors proved the significantly reduced immunogenicity of the modified FVIII variant by revealing less proliferation of T helper cells to this variant than to the unmodified FVIII. CONCLUSION In silico predictions resulted in a partially deimmunized FVIII. This FVIII is fully functional and was demonstrated to be less immunogenic in in vitro testing. This approach may result in a reduction of the inhibitor risk for patients with hemophilia A.
Collapse
Affiliation(s)
| | | | - Anne S. De Groot
- EpiVax, Inc.ProvidenceRhode IslandUSA
- Center for Vaccines and ImmunologyUniversity of GeorgiaAthensGeorgiaUSA
| | | | | | | |
Collapse
|
32
|
Heinrich MA, Mostafa AMRH, Morton JP, Hawinkels LJAC, Prakash J. Translating complexity and heterogeneity of pancreatic tumor: 3D in vitro to in vivo models. Adv Drug Deliv Rev 2021; 174:265-293. [PMID: 33895214 DOI: 10.1016/j.addr.2021.04.018] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 04/16/2021] [Accepted: 04/19/2021] [Indexed: 02/08/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an extremely aggressive type of cancer with an overall survival rate of less than 7-8%, emphasizing the need for novel effective therapeutics against PDAC. However only a fraction of therapeutics which seemed promising in the laboratory environment will eventually reach the clinic. One of the main reasons behind this low success rate is the complex tumor microenvironment (TME) of PDAC, a highly fibrotic and dense stroma surrounding tumor cells, which supports tumor progression as well as increases the resistance against the treatment. In particular, the growing understanding of the PDAC TME points out a different challenge in the development of efficient therapeutics - a lack of biologically relevant in vitro and in vivo models that resemble the complexity and heterogeneity of PDAC observed in patients. The purpose and scope of this review is to provide an overview of the recent developments in different in vitro and in vivo models, which aim to recapitulate the complexity of PDAC in a laboratory environment, as well to describe how 3D in vitro models can be integrated into drug development pipelines that are already including sophisticated in vivo models. Hereby a special focus will be given on the complexity of in vivo models and the challenges in vitro models face to reach the same levels of complexity in a controllable manner. First, a brief introduction of novel developments in two dimensional (2D) models and ex vivo models is provided. Next, recent developments in three dimensional (3D) in vitro models are described ranging from spheroids, organoids, scaffold models, bioprinted models to organ-on-chip models including a discussion on advantages and limitations for each model. Furthermore, we will provide a detailed overview on the current PDAC in vivo models including chemically-induced models, syngeneic and xenogeneic models, highlighting hetero- and orthotopic, patient-derived tissues (PDX) models, and genetically engineered mouse models. Finally, we will provide a discussion on overall limitations of both, in vitro and in vivo models, and discuss necessary steps to overcome these limitations to reach an efficient drug development pipeline, as well as discuss possibilities to include novel in silico models in the process.
Collapse
Affiliation(s)
- Marcel A Heinrich
- Department of Biomaterials Science and Technology, Section Targeted Therapeutics, Technical Medical Centre, University of Twente, 7500AE Enschede, the Netherlands
| | - Ahmed M R H Mostafa
- Department of Biomaterials Science and Technology, Section Targeted Therapeutics, Technical Medical Centre, University of Twente, 7500AE Enschede, the Netherlands
| | - Jennifer P Morton
- Cancer Research UK, Beatson Institute, Garscube Estate, Switchback Rd, Glasgow G61 1BD, UK; Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Rd, Glasgow G61 1QH, UK
| | - Lukas J A C Hawinkels
- Department of Gastroenterology-Hepatology, Leiden University Medical Centre, PO-box 9600, 2300 RC Leiden, the Netherlands
| | - Jai Prakash
- Department of Biomaterials Science and Technology, Section Targeted Therapeutics, Technical Medical Centre, University of Twente, 7500AE Enschede, the Netherlands.
| |
Collapse
|
33
|
Garcia-Sanchez C, Casillas-Abundis MA, Pinelli DF, Tambur AR, Hod-Dvorai R. Impact of SIRPα polymorphism on transplant outcomes in HLA-identical living donor kidney transplantation. Clin Transplant 2021; 35:e14406. [PMID: 34180101 DOI: 10.1111/ctr.14406] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 06/09/2021] [Accepted: 06/21/2021] [Indexed: 12/12/2022]
Abstract
Signal-regulatory protein α (SIRPα), a polymorphic inhibitory membrane-bound receptor, and its ligand CD47 have recently been implicated in the modulation of innate immune allorecognition in murine models. Here, we investigate the potential impact of SIRPα donor-recipient mismatches on graft outcomes in human kidney transplantation. To eliminate the specific role of HLA-matching in alloresponse, we genotyped the two most common variants of SIRPα in a cohort of 55 HLA-identical, biologically-related, donor-recipient pairs. 69% of pairs were SIRPα identical. No significant differences were found between donor-recipient SIRPα-mismatch status and T cell-mediated rejection/borderline changes (25.8% vs. 25%) or slow graft function (15.8% vs. 17.6%). A trend towards more graft failure (GF) (23.5% vs. 5.3%, P = .06), interstitial inflammation (50% vs. 23%, P = .06) and significant changes in peritubular capillaritis (ptc) (25% vs. 0%, P = .02) were observed in the SIRPα-mismatched group. Unexpectedly, graft-versus-host (GVH) SIRPα-mismatched pairs exhibited higher rates of GF and tubulitis (38% vs. 5%, P = .031 and .61 ± .88 vs. 0, P = .019; respectively). Whether the higher prevalence of ptc in SIRPα-mismatched recipients and the higher rates of GF in GVH SIRPα-mismatched pairs represent a potential role for SIRPα in linking innate immunity and alloimmune rejection requires further investigation in larger cohorts.
Collapse
Affiliation(s)
- Cynthia Garcia-Sanchez
- Transplant Immunology Laboratory, Comprehensive Transplant Center, Northwestern University, Chicago, Illinois, USA
| | - M Aurora Casillas-Abundis
- Transplant Immunology Laboratory, Comprehensive Transplant Center, Northwestern University, Chicago, Illinois, USA
| | - David F Pinelli
- Transplant Immunology Laboratory, Comprehensive Transplant Center, Northwestern University, Chicago, Illinois, USA
| | - Anat R Tambur
- Transplant Immunology Laboratory, Comprehensive Transplant Center, Northwestern University, Chicago, Illinois, USA
| | - Reut Hod-Dvorai
- Pathology Department, SUNY Upstate Medical University, Syracuse, New York, USA
| |
Collapse
|
34
|
Jin KT, Du WL, Lan HR, Liu YY, Mao CS, Du JL, Mou XZ. Development of humanized mouse with patient-derived xenografts for cancer immunotherapy studies: A comprehensive review. Cancer Sci 2021; 112:2592-2606. [PMID: 33938090 PMCID: PMC8253285 DOI: 10.1111/cas.14934] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 04/22/2021] [Accepted: 04/24/2021] [Indexed: 02/06/2023] Open
Abstract
Immunotherapy has revolutionized cancer treatment, however, not all tumor types and patients are completely responsive to this approach. Establishing predictive pre-clinical models would allow for more accurate and practical immunotherapeutic drug development. Mouse models are extensively used as in vivo system for biomedical research. However, due to the significant differences between rodents and human, it is impossible to translate most of the findings from mouse models to human. Pharmacological development and advancing personalized medicine using patient-derived xenografts relies on producing mouse models in which murine cells and genes are substituted with their human equivalent. Humanized mice (HM) provide a suitable platform to evaluate xenograft growth in the context of a human immune system. In this review, we discussed recent advances in the generation and application of HM models. We also reviewed new insights into the basic mechanisms, pre-clinical evaluation of onco-immunotherapies, current limitations in the application of these models as well as available improvement strategies. Finally, we pointed out some issues for future studies.
Collapse
Affiliation(s)
- Ke-Tao Jin
- Department of Colorectal Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Wen-Lin Du
- Key Laboratory of Gastroenterology of Zhejiang Province, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China.,Clinical Research Institute, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China
| | - Huan-Rong Lan
- Department of Breast and Thyroid Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Yu-Yao Liu
- Department of Colorectal Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Chun-Sen Mao
- Department of Colorectal Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Jin-Lin Du
- Department of Colorectal Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Xiao-Zhou Mou
- Clinical Research Institute, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China
| |
Collapse
|
35
|
Kelley SM, Ravichandran KS. Putting the brakes on phagocytosis: "don't-eat-me" signaling in physiology and disease. EMBO Rep 2021; 22:e52564. [PMID: 34041845 DOI: 10.15252/embr.202152564] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/12/2021] [Accepted: 04/29/2021] [Indexed: 12/12/2022] Open
Abstract
Timely removal of dying or pathogenic cells by phagocytes is essential to maintaining host homeostasis. Phagocytes execute the clearance process with high fidelity while sparing healthy neighboring cells, and this process is at least partially regulated by the balance of "eat-me" and "don't-eat-me" signals expressed on the surface of host cells. Upon contact, eat-me signals activate "pro-phagocytic" receptors expressed on the phagocyte membrane and signal to promote phagocytosis. Conversely, don't-eat-me signals engage "anti-phagocytic" receptors to suppress phagocytosis. We review the current knowledge of don't-eat-me signaling in normal physiology and disease contexts where aberrant don't-eat-me signaling contributes to pathology.
Collapse
Affiliation(s)
- Shannon M Kelley
- Center for Cell Clearance, University of Virginia, Charlottesville, VA, USA.,Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA, USA
| | - Kodi S Ravichandran
- Center for Cell Clearance, University of Virginia, Charlottesville, VA, USA.,Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA, USA.,VIB-UGent Center for Inflammation Research, Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| |
Collapse
|
36
|
Shapiro MR, Thirawatananond P, Peters L, Sharp RC, Ogundare S, Posgai AL, Perry DJ, Brusko TM. De-coding genetic risk variants in type 1 diabetes. Immunol Cell Biol 2021; 99:496-508. [PMID: 33483996 PMCID: PMC8119379 DOI: 10.1111/imcb.12438] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 01/08/2021] [Accepted: 01/20/2021] [Indexed: 12/13/2022]
Abstract
The conceptual basis for a genetic predisposition underlying the risk for developing type 1 diabetes (T1D) predates modern human molecular genetics. Over half of the genetic risk has been attributed to the human leukocyte antigen (HLA) class II gene region and to the insulin (INS) gene locus - both thought to confer direction of autoreactivity and tissue specificity. Notwithstanding, questions still remain regarding the functional contributions of a vast array of minor polygenic risk variants scattered throughout the genome that likely influence disease heterogeneity and clinical outcomes. Herein, we summarize the available literature related to the T1D-associated coding variants defined at the time of this review, for the genes PTPN22, IFIH1, SH2B3, CD226, TYK2, FUT2, SIRPG, CTLA4, CTSH and UBASH3A. Data from genotype-selected human cohorts are summarized, and studies from the non-obese diabetic (NOD) mouse are presented to describe the functional impact of these variants in relation to innate and adaptive immunity as well as to β-cell fragility, with expression profiles in tissues and peripheral blood highlighted. The contribution of each variant to progression through T1D staging, including environmental interactions, are discussed with consideration of how their respective protein products may serve as attractive targets for precision medicine-based therapeutics to prevent or suspend the development of T1D.
Collapse
Affiliation(s)
- Melanie R Shapiro
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, Diabetes Institute, University of Florida, Gainesville, FL, 32610, USA
| | - Puchong Thirawatananond
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, Diabetes Institute, University of Florida, Gainesville, FL, 32610, USA
| | - Leeana Peters
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, Diabetes Institute, University of Florida, Gainesville, FL, 32610, USA
| | - Robert C Sharp
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, Diabetes Institute, University of Florida, Gainesville, FL, 32610, USA
| | - Similoluwa Ogundare
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, Diabetes Institute, University of Florida, Gainesville, FL, 32610, USA
| | - Amanda L Posgai
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, Diabetes Institute, University of Florida, Gainesville, FL, 32610, USA
| | - Daniel J Perry
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, Diabetes Institute, University of Florida, Gainesville, FL, 32610, USA
| | - Todd M Brusko
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, Diabetes Institute, University of Florida, Gainesville, FL, 32610, USA
- Department of Pediatrics, College of Medicine, Diabetes Institute, University of Florida, Gainesville, FL, 32610, USA
| |
Collapse
|
37
|
Terahara K, Iwabuchi R, Tsunetsugu-Yokota Y. Perspectives on Non-BLT Humanized Mouse Models for Studying HIV Pathogenesis and Therapy. Viruses 2021; 13:v13050776. [PMID: 33924786 PMCID: PMC8145733 DOI: 10.3390/v13050776] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/23/2021] [Accepted: 04/26/2021] [Indexed: 02/07/2023] Open
Abstract
A variety of humanized mice, which are reconstituted only with human hematopoietic stem cells (HSC) or with fetal thymus and HSCs, have been developed and widely utilized as in vivo animal models of HIV-1 infection. The models represent some aspects of HIV-mediated pathogenesis in humans and are useful for the evaluation of therapeutic regimens. However, there are several limitations in these models, including their incomplete immune responses and poor distribution of human cells to the secondary lymphoid tissues. These limitations are common in many humanized mouse models and are critical issues that need to be addressed. As distinct defects exist in each model, we need to be cautious about the experimental design and interpretation of the outcomes obtained using humanized mice. Considering this point, we mainly characterize the current conventional humanized mouse reconstituted only with HSCs and describe past achievements in this area, as well as the potential contributions of the humanized mouse models for the study of HIV pathogenesis and therapy. We also discuss the use of various technologies to solve the current problems. Humanized mice will contribute not only to the pre-clinical evaluation of anti-HIV regimens, but also to a deeper understanding of basic aspects of HIV biology.
Collapse
Affiliation(s)
- Kazutaka Terahara
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo 162-8640, Japan; (K.T.); (R.I.)
| | - Ryutaro Iwabuchi
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo 162-8640, Japan; (K.T.); (R.I.)
- Department of Life Science and Medical Bioscience, Waseda University, Tokyo 162-8480, Japan
| | - Yasuko Tsunetsugu-Yokota
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo 162-8640, Japan; (K.T.); (R.I.)
- Department of Medical Technology, School of Human Sciences, Tokyo University of Technology, Tokyo 144-8535, Japan
- Correspondence: or ; Tel.: +81-3-6424-2223
| |
Collapse
|
38
|
Kaur S, Bronson SM, Pal-Nath D, Miller TW, Soto-Pantoja DR, Roberts DD. Functions of Thrombospondin-1 in the Tumor Microenvironment. Int J Mol Sci 2021; 22:4570. [PMID: 33925464 PMCID: PMC8123789 DOI: 10.3390/ijms22094570] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 04/15/2021] [Accepted: 04/23/2021] [Indexed: 12/13/2022] Open
Abstract
The identification of thrombospondin-1 as an angiogenesis inhibitor in 1990 prompted interest in its role in cancer biology and potential as a therapeutic target. Decreased thrombospondin-1 mRNA and protein expression are associated with progression in several cancers, while expression by nonmalignant cells in the tumor microenvironment and circulating levels in cancer patients can be elevated. THBS1 is not a tumor suppressor gene, but the regulation of its expression in malignant cells by oncogenes and tumor suppressor genes mediates some of their effects on carcinogenesis, tumor progression, and metastasis. In addition to regulating angiogenesis and perfusion of the tumor vasculature, thrombospondin-1 limits antitumor immunity by CD47-dependent regulation of innate and adaptive immune cells. Conversely, thrombospondin-1 is a component of particles released by immune cells that mediate tumor cell killing. Thrombospondin-1 differentially regulates the sensitivity of malignant and nonmalignant cells to genotoxic stress caused by radiotherapy and chemotherapy. The diverse activities of thrombospondin-1 to regulate autophagy, senescence, stem cell maintenance, extracellular vesicle function, and metabolic responses to ischemic and genotoxic stress are mediated by several cell surface receptors and by regulating the functions of several secreted proteins. This review highlights progress in understanding thrombospondin-1 functions in cancer and the challenges that remain in harnessing its therapeutic potential.
Collapse
Affiliation(s)
- Sukhbir Kaur
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA; (S.K.); (D.P.-N.)
| | - Steven M. Bronson
- Department of Internal Medicine, Section of Molecular Medicine, Comprehensive Cancer Center, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA;
| | - Dipasmita Pal-Nath
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA; (S.K.); (D.P.-N.)
| | - Thomas W. Miller
- Centre de Recherche en Cancérologie de Marseille, Institut Paoli-Calmettes, 13273 Marseille, France
| | - David R. Soto-Pantoja
- Department of Surgery and Department of Cancer Biology, Comprehensive Cancer Center, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA
| | - David D. Roberts
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA; (S.K.); (D.P.-N.)
| |
Collapse
|
39
|
Central human B cell tolerance manifests with a distinctive cell phenotype and is enforced via CXCR4 signaling in hu-mice. Proc Natl Acad Sci U S A 2021; 118:2021570118. [PMID: 33850015 DOI: 10.1073/pnas.2021570118] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Central B cell tolerance, the process restricting the development of many newly generated autoreactive B cells, has been intensely investigated in mouse cells while studies in humans have been hampered by the inability to phenotypically distinguish autoreactive and nonautoreactive immature B cell clones and the difficulty in accessing fresh human bone marrow samples. Using a human immune system mouse model in which all human Igκ+ B cells undergo central tolerance, we discovered that human autoreactive immature B cells exhibit a distinctive phenotype that includes lower activation of ERK and differential expression of CD69, CD81, CXCR4, and other glycoproteins. Human B cells exhibiting these characteristics were observed in fresh human bone marrow tissue biopsy specimens, although differences in marker expression were smaller than in the humanized mouse model. Furthermore, the expression of these markers was slightly altered in autoreactive B cells of humanized mice engrafted with some human immune systems genetically predisposed to autoimmunity. Finally, by treating mice and human immune system mice with a pharmacologic antagonist, we show that signaling by CXCR4 is necessary to prevent both human and mouse autoreactive B cell clones from egressing the bone marrow, indicating that CXCR4 functionally contributes to central B cell tolerance.
Collapse
|
40
|
Marín-Jiménez JA, Capasso A, Lewis MS, Bagby SM, Hartman SJ, Shulman J, Navarro NM, Yu H, Rivard CJ, Wang X, Barkow JC, Geng D, Kar A, Yingst A, Tufa DM, Dolan JT, Blatchford PJ, Freed BM, Torres RM, Davila E, Slansky JE, Pelanda R, Eckhardt SG, Messersmith WA, Diamond JR, Lieu CH, Verneris MR, Wang JH, Kiseljak-Vassiliades K, Pitts TM, Lang J. Testing Cancer Immunotherapy in a Human Immune System Mouse Model: Correlating Treatment Responses to Human Chimerism, Therapeutic Variables and Immune Cell Phenotypes. Front Immunol 2021; 12:607282. [PMID: 33854497 PMCID: PMC8040953 DOI: 10.3389/fimmu.2021.607282] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 03/04/2021] [Indexed: 01/22/2023] Open
Abstract
Over the past decade, immunotherapies have revolutionized the treatment of cancer. Although the success of immunotherapy is remarkable, it is still limited to a subset of patients. More than 1500 clinical trials are currently ongoing with a goal of improving the efficacy of immunotherapy through co-administration of other agents. Preclinical, small-animal models are strongly desired to increase the pace of scientific discovery, while reducing the cost of combination drug testing in humans. Human immune system (HIS) mice are highly immune-deficient mouse recipients rtpeconstituted with human hematopoietic stem cells. These HIS-mice are capable of growing human tumor cell lines and patient-derived tumor xenografts. This model allows rapid testing of multiple, immune-related therapeutics for tumors originating from unique clinical samples. Using a cord blood-derived HIS-BALB/c-Rag2nullIl2rγnullSIRPαNOD (BRGS) mouse model, we summarize our experiments testing immune checkpoint blockade combinations in these mice bearing a variety of human tumors, including breast, colorectal, pancreatic, lung, adrenocortical, melanoma and hematological malignancies. We present in-depth characterization of the kinetics and subsets of the HIS in lymph and non-lymph organs and relate these to protocol development and immune-related treatment responses. Furthermore, we compare the phenotype of the HIS in lymph tissues and tumors. We show that the immunotype and amount of tumor infiltrating leukocytes are widely-variable and that this phenotype is tumor-dependent in the HIS-BRGS model. We further present flow cytometric analyses of immune cell subsets, activation state, cytokine production and inhibitory receptor expression in peripheral lymph organs and tumors. We show that responding tumors bear human infiltrating T cells with a more inflammatory signature compared to non-responding tumors, similar to reports of "responding" patients in human immunotherapy clinical trials. Collectively these data support the use of HIS mice as a preclinical model to test combination immunotherapies for human cancers, if careful attention is taken to both protocol details and data analysis.
Collapse
Affiliation(s)
- Juan A. Marín-Jiménez
- Department of Medical Oncology, Catalan Institute of Oncology (ICO-L’Hospitalet), Barcelona, Spain
| | - Anna Capasso
- Department of Oncology, Livestrong Cancer Institutes, Dell Medical School, University of Texas at Austin, Austin, TX, United States
| | - Matthew S. Lewis
- Department of Immunology and Microbiology, School of Medicine, University of Colorado, Aurora, CO, United States
| | - Stacey M. Bagby
- Division of Medical Oncology, School of Medicine, University of Colorado, Aurora, CO, United States
| | - Sarah J. Hartman
- Division of Medical Oncology, School of Medicine, University of Colorado, Aurora, CO, United States
| | - Jeremy Shulman
- Department of Immunology and Microbiology, School of Medicine, University of Colorado, Aurora, CO, United States
| | - Natalie M. Navarro
- Division of Medical Oncology, School of Medicine, University of Colorado, Aurora, CO, United States
| | - Hui Yu
- Division of Medical Oncology, School of Medicine, University of Colorado, Aurora, CO, United States
- University of Colorado Cancer Center, Aurora, CO, United States
| | - Chris J. Rivard
- Division of Medical Oncology, School of Medicine, University of Colorado, Aurora, CO, United States
- University of Colorado Cancer Center, Aurora, CO, United States
| | - Xiaoguang Wang
- Department of Immunology and Microbiology, School of Medicine, University of Colorado, Aurora, CO, United States
| | - Jessica C. Barkow
- Department of Immunology and Microbiology, School of Medicine, University of Colorado, Aurora, CO, United States
| | - Degui Geng
- Division of Medical Oncology, School of Medicine, University of Colorado, Aurora, CO, United States
| | - Adwitiya Kar
- Division of Endocrinology, School of Medicine, University of Colorado, Aurora, CO, United States
| | - Ashley Yingst
- Department of Pediatrics, School of Medicine, University of Colorado, Aurora, CO, United States
| | - Dejene M. Tufa
- Department of Pediatrics, School of Medicine, University of Colorado, Aurora, CO, United States
| | - James T. Dolan
- Rocky Vista College of Osteopathic Medicine – OMS3, Rocky Vista University, Parker, CO, United States
| | - Patrick J. Blatchford
- Department of Biostatistics and Informatics, Colorado School of Public Health, University of Colorado Denver, Aurora, CO, United States
| | - Brian M. Freed
- Department of Immunology and Microbiology, School of Medicine, University of Colorado, Aurora, CO, United States
- Division of Allergy and Clinical Immunology, School of Medicine, University of Colorado, Aurora, CO, United States
| | - Raul M. Torres
- Department of Immunology and Microbiology, School of Medicine, University of Colorado, Aurora, CO, United States
- University of Colorado Cancer Center, Aurora, CO, United States
| | - Eduardo Davila
- Division of Medical Oncology, School of Medicine, University of Colorado, Aurora, CO, United States
- University of Colorado Cancer Center, Aurora, CO, United States
| | - Jill E. Slansky
- Department of Immunology and Microbiology, School of Medicine, University of Colorado, Aurora, CO, United States
- University of Colorado Cancer Center, Aurora, CO, United States
| | - Roberta Pelanda
- Department of Immunology and Microbiology, School of Medicine, University of Colorado, Aurora, CO, United States
- University of Colorado Cancer Center, Aurora, CO, United States
| | - S. Gail Eckhardt
- Department of Oncology, Livestrong Cancer Institutes, Dell Medical School, University of Texas at Austin, Austin, TX, United States
| | - Wells A. Messersmith
- Division of Medical Oncology, School of Medicine, University of Colorado, Aurora, CO, United States
- University of Colorado Cancer Center, Aurora, CO, United States
| | - Jennifer R. Diamond
- Division of Medical Oncology, School of Medicine, University of Colorado, Aurora, CO, United States
- University of Colorado Cancer Center, Aurora, CO, United States
| | - Christopher H. Lieu
- Division of Medical Oncology, School of Medicine, University of Colorado, Aurora, CO, United States
| | - Michael R. Verneris
- Department of Pediatrics, School of Medicine, University of Colorado, Aurora, CO, United States
| | - Jing H. Wang
- Department of Immunology and Microbiology, School of Medicine, University of Colorado, Aurora, CO, United States
- University of Colorado Cancer Center, Aurora, CO, United States
| | - Katja Kiseljak-Vassiliades
- University of Colorado Cancer Center, Aurora, CO, United States
- Division of Endocrinology, School of Medicine, University of Colorado, Aurora, CO, United States
| | - Todd M. Pitts
- Division of Medical Oncology, School of Medicine, University of Colorado, Aurora, CO, United States
- University of Colorado Cancer Center, Aurora, CO, United States
| | - Julie Lang
- Department of Immunology and Microbiology, School of Medicine, University of Colorado, Aurora, CO, United States
| |
Collapse
|
41
|
Gillgrass A, Wessels JM, Yang JX, Kaushic C. Advances in Humanized Mouse Models to Improve Understanding of HIV-1 Pathogenesis and Immune Responses. Front Immunol 2021; 11:617516. [PMID: 33746940 PMCID: PMC7973037 DOI: 10.3389/fimmu.2020.617516] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 12/30/2020] [Indexed: 12/15/2022] Open
Abstract
Although antiretroviral therapy has transformed human immunodeficiency virus-type 1 (HIV-1) from a deadly infection into a chronic disease, it does not clear the viral reservoir, leaving HIV-1 as an uncurable infection. Currently, 1.2 million new HIV-1 infections occur globally each year, with little decrease over many years. Therefore, additional research is required to advance the current state of HIV management, find potential therapeutic strategies, and further understand the mechanisms of HIV pathogenesis and prevention strategies. Non-human primates (NHP) have been used extensively in HIV research and have provided critical advances within the field, but there are several issues that limit their use. Humanized mouse (Hu-mouse) models, or immunodeficient mice engrafted with human immune cells and/or tissues, provide a cost-effective and practical approach to create models for HIV research. Hu-mice closely parallel multiple aspects of human HIV infection and disease progression. Here, we highlight how innovations in Hu-mouse models have advanced HIV-1 research in the past decade. We discuss the effect of different background strains of mice, of modifications on the reconstitution of the immune cells, and the pros and cons of different human cells and/or tissue engraftment methods, on the ability to examine HIV-1 infection and immune response. Finally, we consider the newest advances in the Hu-mouse models and their potential to advance research in emerging areas of mucosal infections, understand the role of microbiota and the complex issues in HIV-TB co-infection. These innovations in Hu-mouse models hold the potential to significantly enhance mechanistic research to develop novel strategies for HIV prevention and therapeutics.
Collapse
Affiliation(s)
- Amy Gillgrass
- Department of Medicine, McMaster University, Hamilton, ON, Canada
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON, Canada
| | - Jocelyn M. Wessels
- Department of Obstetrics and Gynecology, McMaster University, Hamilton, ON, Canada
| | - Jack X. Yang
- Department of Medicine, McMaster University, Hamilton, ON, Canada
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON, Canada
| | - Charu Kaushic
- Department of Medicine, McMaster University, Hamilton, ON, Canada
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
42
|
Martinov T, McKenna KM, Tan WH, Collins EJ, Kehret AR, Linton JD, Olsen TM, Shobaki N, Rongvaux A. Building the Next Generation of Humanized Hemato-Lymphoid System Mice. Front Immunol 2021; 12:643852. [PMID: 33692812 PMCID: PMC7938325 DOI: 10.3389/fimmu.2021.643852] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 01/27/2021] [Indexed: 12/23/2022] Open
Abstract
Since the late 1980s, mice have been repopulated with human hematopoietic cells to study the fundamental biology of human hematopoiesis and immunity, as well as a broad range of human diseases in vivo. Multiple mouse recipient strains have been developed and protocols optimized to efficiently generate these “humanized” mice. Here, we review three guiding principles that have been applied to the development of the currently available models: (1) establishing tolerance of the mouse host for the human graft; (2) opening hematopoietic niches so that they can be occupied by human cells; and (3) providing necessary support for human hematopoiesis. We then discuss four remaining challenges: (1) human hematopoietic lineages that poorly develop in mice; (2) limited antigen-specific adaptive immunity; (3) absent tolerance of the human immune system for its mouse host; and (4) sub-functional interactions between human immune effectors and target mouse tissues. While major advances are still needed, the current models can already be used to answer specific, clinically-relevant questions and hopefully inform the development of new, life-saving therapies.
Collapse
Affiliation(s)
- Tijana Martinov
- Clinical Research Division, Program in Immunology, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
| | - Kelly M McKenna
- Clinical Research Division, Program in Immunology, Fred Hutchinson Cancer Research Center, Seattle, WA, United States.,Graduate Program in Molecular and Cellular Biology, University of Washington, Seattle, WA, United States.,Medical Scientist Training Program, University of Washington, Seattle, WA, United States
| | - Wei Hong Tan
- Clinical Research Division, Program in Immunology, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
| | - Emily J Collins
- Clinical Research Division, Program in Immunology, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
| | - Allie R Kehret
- Clinical Research Division, Program in Immunology, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
| | - Jonathan D Linton
- Clinical Research Division, Program in Immunology, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
| | - Tayla M Olsen
- Clinical Research Division, Program in Immunology, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
| | - Nour Shobaki
- Clinical Research Division, Program in Immunology, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
| | - Anthony Rongvaux
- Clinical Research Division, Program in Immunology, Fred Hutchinson Cancer Research Center, Seattle, WA, United States.,Department of Immunology, University of Washington, Seattle, WA, United States
| |
Collapse
|
43
|
Humanization of Immunodeficient Animals for the Modeling of Transplantation, Graft Versus Host Disease, and Regenerative Medicine. Transplantation 2021; 104:2290-2306. [PMID: 32068660 PMCID: PMC7590965 DOI: 10.1097/tp.0000000000003177] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The humanization of animals is a powerful tool for the exploration of human disease pathogenesis in biomedical research, as well as for the development of therapeutic interventions with enhanced translational potential. Humanized models enable us to overcome biologic differences that exist between humans and other species, while giving us a platform to study human processes in vivo. To become humanized, an immune-deficient recipient is engrafted with cells, tissues, or organoids. The mouse is the most well studied of these hosts, with a variety of immunodeficient strains available for various specific uses. More recently, efforts have turned to the humanization of other animal species such as the rat, which offers some technical and immunologic advantages over mice. These advances, together with ongoing developments in the incorporation of human transgenes and additional mutations in humanized mouse models, have expanded our opportunities to replicate aspects of human allotransplantation and to assist in the development of immunotherapies. In this review, the immune and tissue humanization of various species is presented with an emphasis on their potential for use as models for allotransplantation, graft versus host disease, and regenerative medicine.
Collapse
|
44
|
Abstract
Supplemental Digital Content is available in the text. Background. Humanized immune system immunodeficient mice have been extremely useful for the in vivo analyses of immune responses in a variety of models, including organ transplantation and graft versus host disease (GVHD) but they have limitations. Rat models are interesting complementary alternatives presenting advantages over mice, such as their size and their active complement compartment. Immunodeficient rats have been generated but human immune responses have not yet been described. Methods. We generated immunodeficient Rat Rag−/− Gamma chain−/− human signal regulatory protein alpha-positive (RRGS) rats combining Rag1 and Il2rg deficiency with the expression of human signal regulatory protein alpha, a negative regulator of macrophage phagocytosis allowing repression of rat macrophages by human CD47-positive cells. We then immune humanized RRGS animals with human peripheral blood mononuclear cells (hPBMCs) to set up a human acute GVHD model. Treatment of GVHD was done with a new porcine antihuman lymphocyte serum active through complement-dependent cytotoxicity. We also established a tumor xenograft rejection model in these hPBMCs immune system RRGS animals by subcutaneous implantation of a human tumor cell line. Results. RRGS animals receiving hPBMCs showed robust and reproducible reconstitution, mainly by T and B cells. A dose-dependent acute GVHD process was observed with progressive weight loss, tissue damage, and death censoring. Antihuman lymphocyte serum (L1S1) antibody completely prevented acute GVHD. In the human tumor xenograft model, detectable tumors were rejected upon hPBMCs injection. Conclusions. hPBMC can be implanted in RRGS animals and elicit acute GVHD or rejection of human tumor cells and these are useful models to test new immunotherapies.
Collapse
|
45
|
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: 17] [Impact Index Per Article: 3.4] [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
|
46
|
Wang H, Sun Y, Zhou X, Chen C, Jiao L, Li W, Gou S, Li Y, Du J, Chen G, Zhai W, Wu Y, Qi Y, Gao Y. CD47/SIRPα blocking peptide identification and synergistic effect with irradiation for cancer immunotherapy. J Immunother Cancer 2020; 8:jitc-2020-000905. [PMID: 33020240 PMCID: PMC7537338 DOI: 10.1136/jitc-2020-000905] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/03/2020] [Indexed: 01/04/2023] Open
Abstract
Background Immunotherapy has achieved remarkable advances via a variety of strategies against tumor cells that evade immune surveillance. As important innate immune cells, macrophages play important roles in maintaining homeostasis, preventing pathogen invasion, resisting tumor cells and promoting adaptive immune response. CD47 is found to be overexpressed on tumor cells and act as a don’t eat me’ signal, which contributes to immune evasion. Macrophages mediated phagocytosis via blockade CD47/SIRPα (signal regulatory protein alpha) interaction was proved to induce effective antitumor immune response. Methods A novel peptide pep-20, specifically targeting CD47 and blocking CD47/SIRPα interaction, was identified via high-throughput phage display library bio-panning. The capability to enhance the macrophage-mediated phagocytosis activities and antitumor effects of pep-20 were investigated. The mechanism of pep-20 to induce T-cell response was explored by ex vivo analysis and confirmed via macrophage depleting strategy. The structure-activity relationship and D-amino acid substitution of pep-20 were also studied. The antitumor effects and mechanism of a proteolysis resistant D-amino acid derivate pep-20-D12 combined with irradiation (IR) were also investigated. Results Pep-20 showed remarkable enhancement of macrophage-mediated phagocytosis to both solid and hematologic tumor cells in vitro, and inhibited tumor growth in immune-competent tumor-bearing mice. Furthermore, pep-20 promoted macrophages to mobilize the antitumor T-cell response with minimal toxicity. Furthermore, systemic administration of the derivate pep-20-D12 showed robust synergistic antitumor efficacy in combination with IR. Conclusion In summary, these results demonstrated that CD47/SIRPα blocking peptides, pep-20 and its derivate, could serve as promising candidates to promote macrophages-mediated phagocytosis and immune response in cancer immunotherapy.
Collapse
Affiliation(s)
- Hongfei Wang
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Yixuan Sun
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Xiuman Zhou
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Chunxia Chen
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Ling Jiao
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Wanqiong Li
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Shanshan Gou
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Yanying Li
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Jiangfeng Du
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Guanyu Chen
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Wenjie Zhai
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Yahong Wu
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Yuanming Qi
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Yanfeng Gao
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, Guangdong, China
| |
Collapse
|
47
|
Rautela J, Surgenor E, Huntington ND. Drug target validation in primary human natural killer cells using CRISPR RNP. J Leukoc Biol 2020; 108:1397-1408. [PMID: 33463756 DOI: 10.1002/jlb.2ma0620-074r] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 06/19/2020] [Accepted: 06/21/2020] [Indexed: 12/13/2022] Open
Abstract
The ability to genetically modify CD8 T cells using viral gene delivery has facilitated the development of next generation of cancer immunotherapies such as chimeric Ag receptor (CAR) T cells engineered to specifically kill tumor cells. Development of immunotherapies targeting NK cells have stalled in part by their resistance to traditional viral gene delivery systems. Here, an efficient approach is described to genetically edit human NK cells by electroporation and CRISPR-Cas9 ribonucleoprotein (RNP) complexes. Electroporation pulse codes and buffer optimization for protein uptake by human NK cells and viability, and the efficiency of this approach over other methods are detailed. To highlight the transformative step this technique will have for NK cell immunotherapy drug discovery, NCR1 and CISH are deleted in primary human NK cells and murine findings are validated on their key roles in regulating NK cell antitumor function.
Collapse
Affiliation(s)
- Jai Rautela
- Molecular Immunology Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.,Department of Medical Biology, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Victoria, Australia
| | - Elliot Surgenor
- Molecular Immunology Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Nicholas D Huntington
- Molecular Immunology Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.,Department of Medical Biology, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
48
|
Saito Y, Shultz LD, Ishikawa F. Understanding Normal and Malignant Human Hematopoiesis Using Next-Generation Humanized Mice. Trends Immunol 2020; 41:706-720. [PMID: 32631635 DOI: 10.1016/j.it.2020.06.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 06/12/2020] [Accepted: 06/14/2020] [Indexed: 12/11/2022]
Abstract
Rodent models for human diseases contribute significantly to understanding human physiology and pathophysiology. However, given the accelerating pace of drug development, there is a crucial need for in vivo preclinical models of human biology and pathology. The humanized mouse is one tool to bridge the gap between traditional animal models and the clinic. The development of immunodeficient mouse strains with high-level engraftment of normal and diseased human immune/hematopoietic cells has made in vivo functional characterization possible. As a patient-derived xenograft (PDX) model, humanized mice functionally correlate putative mechanisms with in vivo behavior and help to reveal pathogenic mechanisms. Combined with single-cell genomics, humanized mice can facilitate functional precision medicine such as risk stratification and individually optimized therapeutic approaches.
Collapse
Affiliation(s)
- Yoriko Saito
- RIKEN Center for Integrative Medical Sciences, Yokohama City, Kanagawa, 230-0045, Japan
| | | | - Fumihiko Ishikawa
- RIKEN Center for Integrative Medical Sciences, Yokohama City, Kanagawa, 230-0045, Japan.
| |
Collapse
|
49
|
Abstract
Immune responses to infectious agents are initiated when a pathogen or its components bind to pattern recognition receptors (PRRs). PRR binding sets off a cascade of events that activates immune responses. We now show that, in addition to activating immune responses, PRR signaling also initiates an immunosuppressive response, probably to limit inflammation. The importance of the current findings is that blockade of immunomodulatory signaling, which is mediated by the upregulation of the CD47 molecule, can lead to enhanced immune responses to any pathogen that triggers PRR signaling. Since most or all pathogens trigger PRRs, CD47 blockade could be used to speed up and strengthen both innate and adaptive immune responses when medically indicated. Such immunotherapy could be done without a requirement for knowing the HLA type of the individual, the specific antigens of the pathogen, or, in the case of bacterial infections, the antimicrobial resistance profile. It is well understood that the adaptive immune response to infectious agents includes a modulating suppressive component as well as an activating component. We now show that the very early innate response also has an immunosuppressive component. Infected cells upregulate the CD47 “don’t eat me” signal, which slows the phagocytic uptake of dying and viable cells as well as downstream antigen-presenting cell (APC) functions. A CD47 mimic that acts as an essential virulence factor is encoded by all poxviruses, but CD47 expression on infected cells was found to be upregulated even by pathogens, including severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), that encode no mimic. CD47 upregulation was revealed to be a host response induced by the stimulation of both endosomal and cytosolic pathogen recognition receptors (PRRs). Furthermore, proinflammatory cytokines, including those found in the plasma of hepatitis C patients, upregulated CD47 on uninfected dendritic cells, thereby linking innate modulation with downstream adaptive immune responses. Indeed, results from antibody-mediated CD47 blockade experiments as well as CD47 knockout mice revealed an immunosuppressive role for CD47 during infections with lymphocytic choriomeningitis virus and Mycobacterium tuberculosis. Since CD47 blockade operates at the level of pattern recognition receptors rather than at a pathogen or antigen-specific level, these findings identify CD47 as a novel potential immunotherapeutic target for the enhancement of immune responses to a broad range of infectious agents.
Collapse
|
50
|
Zhang J, Tan SB, Guo ZG. CD47 decline in pancreatic islet cells promotes macrophage-mediated phagocytosis in type I diabetes. World J Diabetes 2020; 11:239-251. [PMID: 32547698 PMCID: PMC7284019 DOI: 10.4239/wjd.v11.i6.239] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 03/30/2020] [Accepted: 04/19/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Type I diabetes (T1D) is characterized by insulin loss caused by inflammatory cells that excessively infiltrate and destroy the pancreas, resulting in dysregulation of tissue homeostasis, mechanobiological properties, and the immune response. The streptozotocin (STZ)-induced mouse model exhibits multiple features of human T1D and enables mechanistic analysis of disease progression. However, the relationship between the mechanochemical signaling regulation of STZ-induced T1D and macrophage migration and phagocytosis is unclear.
AIM To study the mechanochemical regulation of STZ-induced macrophage response on pancreatic beta islet cells to gain a clearer understanding of T1D.
METHODS We performed experiments using different methods. We stimulated isolated pancreatic beta islet cells with STZ and then tested the macrophage migration and phagocytosis.
RESULTS In this study, we discovered that the integrin-associated surface factor CD47 played a critical role in immune defense in the STZ-induced T1D model by preventing pancreatic beta islet inflammation. In comparison with healthy mice, STZ-treated mice showed decreased levels of CD47 on islet cells and reduced interaction of CD47 with signal regulatory protein α (SIRPα), which negatively regulates macrophage-mediated phagocytosis. This resulted in weakened islet cell immune defense and promoted macrophage migration and phagocytosis of target inflammatory cells. Moreover, lipopolysaccharide-activated human acute monocytic leukemia THP-1 cells also exhibited enhanced phagocytosis in the STZ-treated islets, and the aggressive attack of the inflammatory islets correlated with impaired CD47-SIRPα interactions. In addition, CD47 overexpression rescued the pre-labeled targeted cells.
CONCLUSION This study indicates that CD47 deficiency promotes the migration and phagocytosis of macrophages and provides mechanistic insights into T1D by associating the interactions between membrane structures and inflammatory disease progression.
Collapse
Affiliation(s)
- Jing Zhang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210097, Jiangsu Province, China
- Laboratory of Molecular Gerontology, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, United States
| | - Su-Bee Tan
- National Key Laboratory for Biochemistry, College of Life Sciences, Nanjing University, Nanjing 210093, Jiangsu Province, China
| | - Zhi-Gang Guo
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210097, Jiangsu Province, China
| |
Collapse
|