551
|
Langenau DM, Sweet-Cordero A, Wechsler-Reya R, Dyer MA. Preclinical Models Provide Scientific Justification and Translational Relevance for Moving Novel Therapeutics into Clinical Trials for Pediatric Cancer. Cancer Res 2015; 75:5176-5186. [PMID: 26627009 DOI: 10.1158/0008-5472.can-15-1308] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 06/29/2015] [Indexed: 11/16/2022]
Abstract
Despite improvements in survival rates for children with cancer since the 1960s, progress for many pediatric malignancies has slowed over the past two decades. With the recent advances in our understanding of the genomic landscape of pediatric cancer, there is now enthusiasm for individualized cancer therapy based on genomic profiling of patients' tumors. However, several obstacles to effective personalized cancer therapy remain. For example, relatively little data from prospective clinical trials demonstrate the selective efficacy of molecular-targeted therapeutics based on somatic mutations in the patient's tumor. In this commentary, we discuss recent advances in preclinical testing for pediatric cancer and provide recommendations for providing scientific justification and translational relevance for novel therapeutic combinations for childhood cancer. Establishing rigorous criteria for defining and validating druggable mutations will be essential for the success of ongoing and future clinical genomic trials for pediatric malignancies.
Collapse
Affiliation(s)
- David M Langenau
- Molecular Pathology, Cancer Center, and Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02129.,Harvard Stem Cell Institute, Cambridge MA 02139
| | - Alejandro Sweet-Cordero
- Pediatrics, Stanford University Medical School. 265 Campus Drive, LLSCR Building Rm G2078b. Stanford, CA, 94305
| | - Robert Wechsler-Reya
- Tumor Initiation and Maintenance Program, NCI-Designated Cancer Center, Sanford-Burnham Medical Research Institute, La Jolla, CA 92037
| | - Michael A Dyer
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, Tennessee, 38105, USA.,Howard Hughes Medical Institute, Chevy Chase, Maryland 20815, USA
| |
Collapse
|
552
|
Jiminez JA, Uwiera TC, Douglas Inglis G, Uwiera RRE. Animal models to study acute and chronic intestinal inflammation in mammals. Gut Pathog 2015; 7:29. [PMID: 26561503 PMCID: PMC4641401 DOI: 10.1186/s13099-015-0076-y] [Citation(s) in RCA: 143] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 10/22/2015] [Indexed: 02/06/2023] Open
Abstract
Acute and chronic inflammatory diseases of the intestine impart a significant and negative impact on the health and well-being of human and non-human mammalian animals. Understanding the underlying mechanisms of inflammatory disease is mandatory to develop effective treatment and prevention strategies. As inflammatory disease etiologies are multifactorial, the use of appropriate animal models and associated metrics of disease are essential. In this regard, animal models used alone or in combination to study acute and chronic inflammatory disease of the mammalian intestine paired with commonly used inflammation-inducing agents are reviewed. This includes both chemical and biological incitants of inflammation, and both non-mammalian (i.e. nematodes, insects, and fish) and mammalian (i.e. rodents, rabbits, pigs, ruminants, dogs, and non-human primates) models of intestinal inflammation including germ-free, gnotobiotic, as well as surgical, and genetically modified animals. Importantly, chemical and biological incitants induce inflammation via a multitude of mechanisms, and intestinal inflammation and injury can vary greatly according to the incitant and animal model used, allowing studies to ascertain both long-term and short-term effects of inflammation. Thus, researchers and clinicians should be aware of the relative strengths and limitations of the various animal models used to study acute and chronic inflammatory diseases of the mammalian intestine, and the scope and relevance of outcomes achievable based on this knowledge. The ability to induce inflammation to mimic common human diseases is an important factor of a successful animal model, however other mechanisms of disease such as the amount of infective agent to induce disease, invasion mechanisms, and the effect various physiologic changes can have on inducing damage are also important features. In many cases, the use of multiple animal models in combination with both chemical and biological incitants is necessary to answer the specific question being addressed regarding intestinal disease. Some incitants can induce acute responses in certain animal models while others can be used to induce chronic responses; this review aims to illustrate the strengths and weaknesses in each animal model and to guide the choice of an appropriate acute or chronic incitant to facilitate intestinal disease.
Collapse
Affiliation(s)
- Janelle A. Jiminez
- />Agriculture and Agri-Food Canada Research Centre, Lethbridge, AB Canada
- />Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB Canada
| | - Trina C. Uwiera
- />Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB Canada
| | - G. Douglas Inglis
- />Agriculture and Agri-Food Canada Research Centre, Lethbridge, AB Canada
| | - Richard R. E. Uwiera
- />Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB Canada
| |
Collapse
|
553
|
Beer PA, Eaves CJ. Modeling Normal and Disordered Human Hematopoiesis. Trends Cancer 2015; 1:199-210. [DOI: 10.1016/j.trecan.2015.09.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Revised: 09/09/2015] [Accepted: 09/11/2015] [Indexed: 02/06/2023]
|
554
|
Hartwig H, Silvestre-Roig C, Hendrikse J, Beckers L, Paulin N, Van der Heiden K, Braster Q, Drechsler M, Daemen MJ, Lutgens E, Soehnlein O. Atherosclerotic Plaque Destabilization in Mice: A Comparative Study. PLoS One 2015; 10:e0141019. [PMID: 26492161 PMCID: PMC4619621 DOI: 10.1371/journal.pone.0141019] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 10/02/2015] [Indexed: 11/25/2022] Open
Abstract
Atherosclerosis-associated diseases are the main cause of mortality and morbidity in western societies. The progression of atherosclerosis is a dynamic process evolving from early to advanced lesions that may become rupture-prone vulnerable plaques. Acute coronary syndromes are the clinical manifestation of life-threatening thrombotic events associated with high-risk vulnerable plaques. Hyperlipidemic mouse models have been extensively used in studying the mechanisms controlling initiation and progression of atherosclerosis. However, the understanding of mechanisms leading to atherosclerotic plaque destabilization has been hampered by the lack of proper animal models mimicking this process. Although various mouse models generate atherosclerotic plaques with histological features of human advanced lesions, a consensus model to study atherosclerotic plaque destabilization is still lacking. Hence, we studied the degree and features of plaque vulnerability in different mouse models of atherosclerotic plaque destabilization and find that the model based on the placement of a shear stress modifier in combination with hypercholesterolemia represent with high incidence the most human like lesions compared to the other models.
Collapse
Affiliation(s)
- Helene Hartwig
- Department of Pathology, Academic Medical Center, Amsterdam, The Netherlands
| | - Carlos Silvestre-Roig
- Department of Pathology, Academic Medical Center, Amsterdam, The Netherlands
- Institute for Cardiovascular Prevention (IPEK), LMU Munich, Munich, Germany
| | - Jeffrey Hendrikse
- Department of Pathology, Academic Medical Center, Amsterdam, The Netherlands
| | - Linda Beckers
- Department of Medical Biochemistry, Academic Medical Center, Amsterdam, The Netherlands
| | - Nicole Paulin
- Institute for Cardiovascular Prevention (IPEK), LMU Munich, Munich, Germany
| | - Kim Van der Heiden
- Department of Cardiology, Biomedical Engineering, Erasmus MC, Rotterdam, The Netherlands
| | - Quinte Braster
- Department of Pathology, Academic Medical Center, Amsterdam, The Netherlands
| | - Maik Drechsler
- Department of Pathology, Academic Medical Center, Amsterdam, The Netherlands
- Institute for Cardiovascular Prevention (IPEK), LMU Munich, Munich, Germany
| | - Mat J. Daemen
- Department of Pathology, Academic Medical Center, Amsterdam, The Netherlands
| | - Esther Lutgens
- Institute for Cardiovascular Prevention (IPEK), LMU Munich, Munich, Germany
- Department of Medical Biochemistry, Academic Medical Center, Amsterdam, The Netherlands
| | - Oliver Soehnlein
- Department of Pathology, Academic Medical Center, Amsterdam, The Netherlands
- Institute for Cardiovascular Prevention (IPEK), LMU Munich, Munich, Germany
- German Centre for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich, Germany
- * E-mail:
| |
Collapse
|
555
|
Goyama S, Schibler J, Gasilina A, Shrestha M, Lin S, Link KA, Chen J, Whitman SP, Bloomfield CD, Nicolet D, Assi SA, Ptasinska A, Heidenreich O, Bonifer C, Kitamura T, Nassar NN, Mulloy JC. UBASH3B/Sts-1-CBL axis regulates myeloid proliferation in human preleukemia induced by AML1-ETO. Leukemia 2015; 30:728-39. [PMID: 26449661 DOI: 10.1038/leu.2015.275] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Revised: 09/22/2015] [Accepted: 09/30/2015] [Indexed: 12/24/2022]
Abstract
The t(8;21) rearrangement, which creates the AML1-ETO fusion protein, represents the most common chromosomal translocation in acute myeloid leukemia (AML). Clinical data suggest that CBL mutations are a frequent event in t(8;21) AML, but the role of CBL in AML1-ETO-induced leukemia has not been investigated. In this study, we demonstrate that CBL mutations collaborate with AML1-ETO to expand human CD34+ cells both in vitro and in a xenograft model. CBL depletion by shRNA also promotes the growth of AML1-ETO cells, demonstrating the inhibitory function of endogenous CBL in t(8;21) AML. Mechanistically, loss of CBL function confers hyper-responsiveness to thrombopoietin and enhances STAT5/AKT/ERK/Src signaling in AML1-ETO cells. Interestingly, we found the protein tyrosine phosphatase UBASH3B/Sts-1, which is known to inhibit CBL function, is upregulated by AML1-ETO through transcriptional and miR-9-mediated regulation. UBASH3B/Sts-1 depletion induces an aberrant pattern of CBL phosphorylation and impairs proliferation in AML1-ETO cells. The growth inhibition caused by UBASH3B/Sts-1 depletion can be rescued by ectopic expression of CBL mutants, suggesting that UBASH3B/Sts-1 supports the growth of AML1-ETO cells partly through modulation of CBL function. Our study reveals a role of CBL in restricting myeloid proliferation of human AML1-ETO-induced leukemia, and identifies UBASH3B/Sts-1 as a potential target for pharmaceutical intervention.
Collapse
Affiliation(s)
- S Goyama
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA.,Division of Cellular Therapy, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - J Schibler
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - A Gasilina
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - M Shrestha
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - S Lin
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - K A Link
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - J Chen
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - S P Whitman
- The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - C D Bloomfield
- The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - D Nicolet
- The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA.,Alliance for Clinical Trials in Oncology Statistics and Data Center, Mayo Clinic, Rochester, MN, USA
| | - S A Assi
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - A Ptasinska
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - O Heidenreich
- Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne, UK
| | - C Bonifer
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - T Kitamura
- Division of Cellular Therapy, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - N N Nassar
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - J C Mulloy
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| |
Collapse
|
556
|
Kuzu OF, Nguyen FD, Noory MA, Sharma A. Current State of Animal (Mouse) Modeling in Melanoma Research. CANCER GROWTH AND METASTASIS 2015; 8:81-94. [PMID: 26483610 PMCID: PMC4597587 DOI: 10.4137/cgm.s21214] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Revised: 08/10/2015] [Accepted: 08/17/2015] [Indexed: 11/16/2022]
Abstract
Despite the considerable progress in understanding the biology of human cancer and technological advancement in drug discovery, treatment failure remains an inevitable outcome for most cancer patients with advanced diseases, including melanoma. Despite FDA-approved BRAF-targeted therapies for advanced stage melanoma showed a great deal of promise, development of rapid resistance limits the success. Hence, the overall success rate of melanoma therapy still remains to be one of the worst compared to other malignancies. Advancement of next-generation sequencing technology allowed better identification of alterations that trigger melanoma development. As development of successful therapies strongly depends on clinically relevant preclinical models, together with the new findings, more advanced melanoma models have been generated. In this article, besides traditional mouse models of melanoma, we will discuss recent ones, such as patient-derived tumor xenografts, topically inducible BRAF mouse model and RCAS/TVA-based model, and their advantages as well as limitations. Although mouse models of melanoma are often criticized as poor predictors of whether an experimental drug would be an effective treatment, development of new and more relevant models could circumvent this problem in the near future.
Collapse
Affiliation(s)
- Omer F Kuzu
- Department of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Felix D Nguyen
- The University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Mohammad A Noory
- Department of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Arati Sharma
- Department of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, PA, USA
| |
Collapse
|
557
|
Pearson JA, Wong FS, Wen L. The importance of the Non Obese Diabetic (NOD) mouse model in autoimmune diabetes. J Autoimmun 2015; 66:76-88. [PMID: 26403950 DOI: 10.1016/j.jaut.2015.08.019] [Citation(s) in RCA: 196] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Accepted: 08/26/2015] [Indexed: 02/06/2023]
Abstract
Type 1 Diabetes (T1D) is an autoimmune disease characterized by the pancreatic infiltration of immune cells resulting in T cell-mediated destruction of the insulin-producing beta cells. The successes of the Non-Obese Diabetic (NOD) mouse model have come in multiple forms including identifying key genetic and environmental risk factors e.g. Idd loci and effects of microorganisms including the gut microbiota, respectively, and how they may contribute to disease susceptibility and pathogenesis. Furthermore, the NOD model also provides insights into the roles of the innate immune cells as well as the B cells in contributing to the T cell-mediated disease. Unlike many autoimmune disease models, the NOD mouse develops spontaneous disease and has many similarities to human T1D. Through exploiting these similarities many targets have been identified for immune-intervention strategies. Although many of these immunotherapies did not have a significant impact on human T1D, they have been shown to be effective in the NOD mouse in early stage disease, which is not equivalent to trials in newly-diagnosed patients with diabetes. However, the continued development of humanized NOD mice would enable further clinical developments, bringing T1D research to a new translational level. Therefore, it is the aim of this review to discuss the importance of the NOD model in identifying the roles of the innate immune system and the interaction with the gut microbiota in modifying diabetes susceptibility. In addition, the role of the B cells will also be discussed with new insights gained through B cell depletion experiments and the impact on translational developments. Finally, this review will also discuss the future of the NOD mouse and the development of humanized NOD mice, providing novel insights into human T1D.
Collapse
Affiliation(s)
- James A Pearson
- Section of Endocrinology, School of Medicine, Yale University, New Haven, CT, USA
| | - F Susan Wong
- Diabetes Research Group, Institute of Molecular & Experimental Medicine, School of Medicine, Cardiff University, Wales, UK
| | - Li Wen
- Section of Endocrinology, School of Medicine, Yale University, New Haven, CT, USA.
| |
Collapse
|
558
|
Radigan KA, Misharin AV, Chi M, Budinger GRS. Modeling human influenza infection in the laboratory. Infect Drug Resist 2015; 8:311-20. [PMID: 26357484 PMCID: PMC4560508 DOI: 10.2147/idr.s58551] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Influenza is the leading cause of death from an infectious cause. Because of its clinical importance, many investigators use animal models to understand the biologic mechanisms of influenza A virus replication, the immune response to the virus, and the efficacy of novel therapies. This review will focus on the biosafety, biosecurity, and ethical concerns that must be considered in pursuing influenza research, in addition to focusing on the two animal models - mice and ferrets - most frequently used by researchers as models of human influenza infection.
Collapse
Affiliation(s)
| | - Alexander V Misharin
- Division of Rheumatology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Monica Chi
- Division of Pulmonary and Critical Care Medicine, Chicago, IL, USA
| | | |
Collapse
|
559
|
Riese P, Trittel S, Schulze K, Guzmán CA. Rodents as pre-clinical models for predicting vaccine performance in humans. Expert Rev Vaccines 2015. [DOI: 10.1586/14760584.2015.1074043] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
| | - Stephanie Trittel
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - Kai Schulze
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - Carlos A Guzmán
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| |
Collapse
|
560
|
Barrett DM, Grupp SA, June CH. Chimeric Antigen Receptor- and TCR-Modified T Cells Enter Main Street and Wall Street. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2015; 195:755-61. [PMID: 26188068 PMCID: PMC4507286 DOI: 10.4049/jimmunol.1500751] [Citation(s) in RCA: 123] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The field of adoptive cell transfer (ACT) is currently comprised of chimeric Ag receptor (CAR)- and TCR-engineered T cells and has emerged from principles of basic immunology to paradigm-shifting clinical immunotherapy. ACT of T cells engineered to express artificial receptors that target cells of choice is an exciting new approach for cancer, and it holds equal promise for chronic infection and autoimmunity. Using principles of synthetic biology, advances in immunology, and genetic engineering have made it possible to generate human T cells that display desired specificities and enhanced functionalities. Clinical trials in patients with advanced B cell leukemias and lymphomas treated with CD19-specific CAR T cells have induced durable remissions in adults and children. The prospects for the widespread availability of engineered T cells have changed dramatically given the recent entry of the pharmaceutical industry to this arena. In this overview, we discuss some of the challenges and opportunities that face the field of ACT.
Collapse
Affiliation(s)
- David M Barrett
- Division of Oncology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA 19104
| | - Stephan A Grupp
- Division of Oncology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA 19104; Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104; and
| | - Carl H June
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104; and Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| |
Collapse
|
561
|
Vercauteren K, de Jong YP, Meuleman P. Animal models for the study of HCV. Curr Opin Virol 2015; 13:67-74. [PMID: 26304554 PMCID: PMC4549803 DOI: 10.1016/j.coviro.2015.04.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Revised: 04/29/2015] [Accepted: 04/30/2015] [Indexed: 12/18/2022]
Abstract
The development and evaluation of effective therapies and vaccines for the hepatitis C virus (HCV) and the study of its interactions with the mammalian host have been hindered for a long time by the absence of suitable small animal models. Immune compromised mouse models that recapitulate the complete HCV life cycle have been useful to investigate many aspects of the HCV life cycle including antiviral interventions. However, HCV has a high propensity to establish persistence and associated histopathological manifestations such as steatosis, fibrosis, cirrhosis and hepatocellular carcinoma (HCC). Better understanding of these processes requires the development of a permissive and fully immunocompetent small animal model. In this review we summarize the in vivo models that are available for the study of HCV.
Collapse
Affiliation(s)
- Koen Vercauteren
- Center for Vaccinology, Dept. of Clinical Chemistry, Microbiology and Immunology, Ghent University, Ghent, Belgium; Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, USA
| | - Ype P de Jong
- Division of Gastroenterology and Hepatology, Weill Cornell Medical College, New York, USA; Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, USA
| | - Philip Meuleman
- Center for Vaccinology, Dept. of Clinical Chemistry, Microbiology and Immunology, Ghent University, Ghent, Belgium.
| |
Collapse
|
562
|
Carpenter RS, Kigerl KA, Marbourg JM, Gaudet AD, Huey D, Niewiesk S, Popovich PG. Traumatic spinal cord injury in mice with human immune systems. Exp Neurol 2015; 271:432-44. [PMID: 26193167 DOI: 10.1016/j.expneurol.2015.07.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Revised: 06/18/2015] [Accepted: 07/13/2015] [Indexed: 01/21/2023]
Abstract
Mouse models have provided key insight into the cellular and molecular control of human immune system function. However, recent data indicate that extrapolating the functional capabilities of the murine immune system into humans can be misleading. Since immune cells significantly affect neuron survival and axon growth and also are required to defend the body against infection, it is important to determine the pathophysiological significance of spinal cord injury (SCI)-induced changes in human immune system function. Research projects using monkeys or humans would be ideal; however, logistical and ethical barriers preclude detailed mechanistic studies in either species. Humanized mice, i.e., immunocompromised mice reconstituted with human immune cells, can help overcome these barriers and can be applied in various experimental conditions that are of interest to the SCI community. Specifically, newborn NOD-SCID-IL2rg(null) (NSG) mice engrafted with human CD34(+) hematopoietic stem cells develop normally without neurological impairment. In this report, new data show that when mice with human immune systems receive a clinically-relevant spinal contusion injury, spontaneous functional recovery is indistinguishable from that achieved after SCI using conventional inbred mouse strains. Moreover, using routine immunohistochemical and flow cytometry techniques, one can easily phenotype circulating human immune cells and document the composition and distribution of these cells in the injured spinal cord. Lesion pathology in humanized mice is typical of mouse contusion injuries, producing a centralized lesion epicenter that becomes occupied by phagocytic macrophages and lymphocytes and enclosed by a dense astrocytic scar. Specific human immune cell types, including three distinct subsets of human monocytes, were readily detected in the blood, spleen and liver. Future studies that aim to understand the functional consequences of manipulating the neuro-immune axis after SCI should consider using the humanized mouse model. Humanized mice represent a powerful tool for improving the translational value of pre-clinical SCI data.
Collapse
Affiliation(s)
- Randall S Carpenter
- Neuroscience Graduate Studies Program, The Ohio State University, Columbus, OH, USA; Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, OH, USA; Department of Neuroscience, The Ohio State University, Columbus, OH, USA
| | - Kristina A Kigerl
- Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, OH, USA; Department of Neuroscience, The Ohio State University, Columbus, OH, USA
| | - Jessica M Marbourg
- Neuroscience Graduate Studies Program, The Ohio State University, Columbus, OH, USA; Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, OH, USA; Department of Neuroscience, The Ohio State University, Columbus, OH, USA
| | - Andrew D Gaudet
- Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, OH, USA; Department of Neuroscience, The Ohio State University, Columbus, OH, USA
| | - Devra Huey
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, USA
| | - Stefan Niewiesk
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, USA
| | - Phillip G Popovich
- Neuroscience Graduate Studies Program, The Ohio State University, Columbus, OH, USA; Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, OH, USA; Department of Neuroscience, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
563
|
Mouse models for core binding factor leukemia. Leukemia 2015; 29:1970-80. [PMID: 26165235 DOI: 10.1038/leu.2015.181] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Revised: 06/03/2015] [Accepted: 06/18/2015] [Indexed: 02/07/2023]
Abstract
RUNX1 and CBFB are among the most frequently mutated genes in human leukemias. Genetic alterations such as chromosomal translocations, copy number variations and point mutations have been widely reported to result in the malfunction of RUNX transcription factors. Leukemias arising from such alterations in RUNX family genes are collectively termed core binding factor (CBF) leukemias. Although adult CBF leukemias generally are considered a favorable risk group as compared with other forms of acute myeloid leukemia, the 5-year survival rate remains low. An improved understanding of the molecular mechanism for CBF leukemia is imperative to uncover novel treatment options. Over the years, retroviral transduction-transplantation assays and transgenic, knockin and knockout mouse models alone or in combination with mutagenesis have been used to study the roles of RUNX alterations in leukemogenesis. Although successful in inducing leukemia, the existing assays and models possess many inherent limitations. A CBF leukemia model which induces leukemia with complete penetrance and short latency would be ideal as a platform for drug discovery. Here, we summarize the currently available mouse models which have been utilized to study CBF leukemias, discuss the advantages and limitations of individual experimental systems, and propose suggestions for improvements of mouse models.
Collapse
|
564
|
Koboziev I, Jones-Hall Y, Valentine JF, Webb CR, Furr KL, Grisham MB. Use of Humanized Mice to Study the Pathogenesis of Autoimmune and Inflammatory Diseases. Inflamm Bowel Dis 2015; 21:1652-73. [PMID: 26035036 PMCID: PMC4466023 DOI: 10.1097/mib.0000000000000446] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Animal models of disease have been used extensively by the research community for the past several decades to better understand the pathogenesis of different diseases and assess the efficacy and toxicity of different therapeutic agents. Retrospective analyses of numerous preclinical intervention studies using mouse models of acute and chronic inflammatory diseases reveal a generalized failure to translate promising interventions or therapeutics into clinically effective treatments in patients. Although several possible reasons have been suggested to account for this generalized failure to translate therapeutic efficacy from the laboratory bench to the patient's bedside, it is becoming increasingly apparent that the mouse immune system is substantially different from the human. Indeed, it is well known that >80 major differences exist between mouse and human immunology; all of which contribute to significant differences in immune system development, activation, and responses to challenges in innate and adaptive immunity. This inconvenient reality has prompted investigators to attempt to humanize the mouse immune system to address important human-specific questions that are impossible to study in patients. The successful long-term engraftment of human hematolymphoid cells in mice would provide investigators with a relatively inexpensive small animal model to study clinically relevant mechanisms and facilitate the evaluation of human-specific therapies in vivo. The discovery that targeted mutation of the IL-2 receptor common gamma chain in lymphopenic mice allows for the long-term engraftment of functional human immune cells has advanced greatly our ability to humanize the mouse immune system. The objective of this review is to present a brief overview of the recent advances that have been made in the development and use of humanized mice with special emphasis on autoimmune and chronic inflammatory diseases. In addition, we discuss the use of these unique mouse models to define the human-specific immunopathological mechanisms responsible for the induction and perpetuation of chronic gut inflammation.
Collapse
Affiliation(s)
- Iurii Koboziev
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, Texas 79430
| | - Yava Jones-Hall
- Department of Comparative Pathobiology, Purdue University College of Veterinary Medicine, West Lafayette, IN 47907-2027
| | - John F. Valentine
- Department of Internal Medicine, Gastroenterology, Hepatology and Nutrition, University of Utah, Salt Lake City, UT 84132-2410
| | - Cynthia Reinoso Webb
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, Texas 79430
| | - Kathryn L. Furr
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, Texas 79430
| | - Matthew B. Grisham
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, Texas 79430
| |
Collapse
|
565
|
Abstract
PURPOSE OF REVIEW Experimental models have contributed enormously to basic immunology. However, the use of reductionist experiments has produced results that are not always successfully translated into the clinic. Recently, incorporation of more realistic clinical parameters in experimental designs has produced new insights relevant to cardiac transplantation. RECENT FINDINGS Experiments in mice have provided crucial insights into the concept that T cell responses to pathogens generate memory cells with cross-reactive specificities for histocompatibility antigens. These memory T cells are resistant to current immunosuppressive strategies. Memory T cells infiltrate grafts within hours after transplantation, and grafts subjected to clinically relevant periods of cold ischemia are more susceptible to injury by this cellular infiltrate. Early immune responses now can be investigated with improved 'humanized' mice. Mice with multiple knock-in genes for human cytokines support development of human monocytes, macrophages and natural killer cells in increased numbers and with better function. SUMMARY Better and more clinically relevant experimental designs are providing animal models tailored to address clinic exigencies.
Collapse
|
566
|
Mac Keon S, Ruiz MS, Gazzaniga S, Wainstok R. Dendritic cell-based vaccination in cancer: therapeutic implications emerging from murine models. Front Immunol 2015; 6:243. [PMID: 26042126 PMCID: PMC4438595 DOI: 10.3389/fimmu.2015.00243] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 05/06/2015] [Indexed: 01/29/2023] Open
Abstract
Dendritic cells (DCs) play a pivotal role in the orchestration of immune responses, and are thus key targets in cancer vaccine design. Since the 2010 FDA approval of the first cancer DC-based vaccine (Sipuleucel-T), there has been a surge of interest in exploiting these cells as a therapeutic option for the treatment of tumors of diverse origin. In spite of the encouraging results obtained in the clinic, many elements of DC-based vaccination strategies need to be optimized. In this context, the use of experimental cancer models can help direct efforts toward an effective vaccine design. This paper reviews recent findings in murine models regarding the antitumoral mechanisms of DC-based vaccination, covering issues related to antigen sources, the use of adjuvants and maturing agents, and the role of DC subsets and their interaction in the initiation of antitumoral immune responses. The summary of such diverse aspects will highlight advantages and drawbacks in the use of murine models, and contribute to the design of successful DC-based translational approaches for cancer treatment.
Collapse
Affiliation(s)
- Soledad Mac Keon
- Laboratorio de Cancerología, Fundación Instituto Leloir, Instituto de Investigaciones Bioquímicas de Buenos Aires IIBBA-CONICET , Buenos Aires , Argentina
| | - María Sol Ruiz
- Centro de Investigaciones Oncológicas, Fundación para la Investigación, Docencia y Prevención del Cáncer (FUCA) , Buenos Aires , Argentina
| | - Silvina Gazzaniga
- Laboratorio de Biología Tumoral, Departamento de Química Biológica IQUIBICEN-CONICET, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires , Buenos Aires , Argentina
| | - Rosa Wainstok
- Laboratorio de Cancerología, Fundación Instituto Leloir, Instituto de Investigaciones Bioquímicas de Buenos Aires IIBBA-CONICET , Buenos Aires , Argentina ; Laboratorio de Biología Tumoral, Departamento de Química Biológica IQUIBICEN-CONICET, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires , Buenos Aires , Argentina
| |
Collapse
|
567
|
Campo B, Vandal O, Wesche DL, Burrows JN. Killing the hypnozoite--drug discovery approaches to prevent relapse in Plasmodium vivax. Pathog Glob Health 2015; 109:107-22. [PMID: 25891812 PMCID: PMC4455353 DOI: 10.1179/2047773215y.0000000013] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The eradication of malaria will only be possible if effective, well-tolerated medicines kill hypnozoites in vivax and ovale malaria, and thus prevent relapses in patients. Despite progress in the 8-aminoquinoline series, with tafenoquine in Phase III showing clear benefits over primaquine, the drug discovery challenge to identify hypnozoiticidal or hypnozoite-activating compounds has been hampered by the dearth of biological tools and assays, which in turn has been limited by the immense scientific and logistical challenges associated with accessing relevant human tissue and sporozoites. This review summarises the existing drug discovery series and approaches concerning the goal to block relapse.
Collapse
Affiliation(s)
- Brice Campo
- Medicines for Malaria Venture, Geneva, Switzerland
| | - Omar Vandal
- The Bill and Melinda Gates Foundation, Seattle, WA, USA
| | - David L. Wesche
- The Bill and Melinda Gates Foundation, Seattle, WA, USA
- Great Lakes Drug Development/Certara, Princeton, NJ, USA
| | | |
Collapse
|
568
|
Xenograft models for normal and malignant stem cells. Blood 2015; 125:2630-40. [DOI: 10.1182/blood-2014-11-570218] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Accepted: 03/04/2015] [Indexed: 12/18/2022] Open
Abstract
Abstract
The model systems available for studying human hematopoiesis, malignant hematopoiesis, and hematopoietic stem cell (HSC) function in vivo have improved dramatically over the last decade, primarily due to improvements in xenograft mouse strains. Several recent reviews have focused on the historic development of immunodeficient mice over the last 2 decades, as well as their use in understanding human HSC and leukemia stem cell (LSC) biology and function in the context of a humanized mouse. However, in the intervening time since these reviews, a number of new mouse models, technical approaches, and scientific advances have been made. In this review, we update the reader on the newest and best models and approaches available for studying human malignant and normal HSCs in immunodeficient mice, including newly developed mice for use in chemotherapy testing and improved techniques for humanizing mice without laborious purification of HSC. We also review some relevant scientific findings from xenograft studies and highlight the continued limitations that confront researchers working with human HSC and LSC in vivo.
Collapse
|
569
|
Morton JJ, Bird G, Keysar SB, Astling DP, Lyons TR, Anderson RT, Glogowska MJ, Estes P, Eagles JR, Le PN, Gan G, McGettigan B, Fernandez P, Padilla-Just N, Varella-Garcia M, Song JI, Bowles DW, Schedin P, Tan AC, Roop DR, Wang XJ, Refaeli Y, Jimeno A. XactMice: humanizing mouse bone marrow enables microenvironment reconstitution in a patient-derived xenograft model of head and neck cancer. Oncogene 2015; 35:290-300. [PMID: 25893296 PMCID: PMC4613815 DOI: 10.1038/onc.2015.94] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2014] [Revised: 02/20/2015] [Accepted: 03/03/2015] [Indexed: 12/14/2022]
Abstract
The limitations of cancer cell lines have led to the development of direct patient derived xenograft (PDX) models. However, the interplay between the implanted human cancer cells and recruited mouse stromal and immune cells alters the tumor microenvironment and limits the value of these models. To overcome these constraints, we have developed a technique to expand human hematopoietic stem and progenitor cells (HSPCs) and use them to reconstitute the radiation-depleted bone marrow of a NOD/SCID/IL2rg−/− (NSG) mouse on which a patient’s tumor is then transplanted (XactMice). The human HSPCs produce immune cells that home into the tumor and help replicate its natural microenvironment. Despite previous passage on nude mice, the expression of epithelial, stromal, and immune genes in XactMice tumors aligns more closely to that of the patient tumor than to those grown in non-humanized mice – an effect partially facilitated by human cytokines expressed by both the HSPC progeny and the tumor cells. The human immune and stromal cells produced in the XactMice can help recapitulate the microenvironment of an implanted xenograft, reverse the initial genetic drift seen after passage on non-humanized mice, and provide a more accurate tumor model to guide patient treatment.
Collapse
Affiliation(s)
- J J Morton
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine
| | - G Bird
- Department of Dermatology, University of Colorado School of Medicine
| | - S B Keysar
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine
| | - D P Astling
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine.,Department of Biostatistics and Informatics, University of Colorado School of Medicine
| | - T R Lyons
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine
| | - R T Anderson
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine
| | - M J Glogowska
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine
| | - P Estes
- Department of Dermatology, University of Colorado School of Medicine
| | - J R Eagles
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine
| | - P N Le
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine
| | - G Gan
- Department of Radiation Oncology, University of Colorado School of Medicine
| | - B McGettigan
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine
| | - P Fernandez
- Department of Pathology, University of Colorado School of Medicine
| | - N Padilla-Just
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine
| | - M Varella-Garcia
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine
| | - J I Song
- Department of Otolaryngology, University of Colorado School of Medicine
| | - D W Bowles
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine
| | - P Schedin
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine
| | - A-C Tan
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine.,Department of Biostatistics and Informatics, University of Colorado School of Medicine
| | - D R Roop
- Department of Dermatology, University of Colorado School of Medicine.,Charles C. Gates Center for Regenerative Medicine and Stem Cell Biology, University of Colorado School of Medicine, Aurora, CO, USA
| | - X-J Wang
- Department of Pathology, University of Colorado School of Medicine.,Charles C. Gates Center for Regenerative Medicine and Stem Cell Biology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Y Refaeli
- Department of Dermatology, University of Colorado School of Medicine.,Charles C. Gates Center for Regenerative Medicine and Stem Cell Biology, University of Colorado School of Medicine, Aurora, CO, USA
| | - A Jimeno
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine.,Department of Otolaryngology, University of Colorado School of Medicine.,Charles C. Gates Center for Regenerative Medicine and Stem Cell Biology, University of Colorado School of Medicine, Aurora, CO, USA
| |
Collapse
|
570
|
Barth H. Hepatitis C virus: Is it time to say goodbye yet? Perspectives and challenges for the next decade. World J Hepatol 2015; 7:725-737. [PMID: 25914773 PMCID: PMC4404378 DOI: 10.4254/wjh.v7.i5.725] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Revised: 12/22/2014] [Accepted: 01/20/2015] [Indexed: 02/06/2023] Open
Abstract
The majority of individuals exposed to hepatitis C virus (HCV) establish a persistent infection, which is a leading cause of chronic liver disease, cirrhosis and hepatocellular carcinoma. Major progress has been made during the past twenty-five years in understanding the HCV life cycle and immune responses against HCV infection. Increasing evidence indicates that host genetic factors can significantly influence the outcome of HCV infection and the response to interferon alpha-based antiviral therapy. The arrival of highly effective and convenient treatment regimens for patients chronically infected with HCV has improved prospects for the eradication of HCV worldwide. Clinical trials are evaluating the best anti-viral drug combination, treatment doses and duration. The new treatments are better-tolerated and have shown success rates of more than 95%. However, the recent breakthrough in HCV treatment raises new questions and challenges, including the identification of HCV-infected patients and to link them to appropriate health care, the high pricing of HCV drugs, the emergence of drug resistance or naturally occurring polymorphism in HCV sequences which can compromise HCV treatment response. Finally, we still do not have a vaccine against HCV. In this concise review, we will highlight the progress made in understanding HCV infection and therapy. We will focus on the most significant unsolved problems and the key future challenges in the management of HCV infection.
Collapse
|
571
|
Fatal autoimmunity in mice reconstituted with human hematopoietic stem cells encoding defective FOXP3. Blood 2015; 125:3886-95. [PMID: 25833964 DOI: 10.1182/blood-2014-12-618363] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 03/23/2015] [Indexed: 01/13/2023] Open
Abstract
Mice reconstituted with a human immune system provide a tractable in vivo model to assess human immune cell function. To date, reconstitution of murine strains with human hematopoietic stem cells (HSCs) from patients with monogenic immune disorders have not been reported. One obstacle precluding the development of immune-disease specific "humanized" mice is that optimal adaptive immune responses in current strains have required implantation of autologous human thymic tissue. To address this issue, we developed a mouse strain that lacks murine major histocompatibility complex class II (MHC II) and instead expresses human leukocyte antigen DR1 (HLA-DR1). These mice displayed improved adaptive immune responses when reconstituted with human HSCs including enhanced T-cell reconstitution, delayed-type hypersensitivity responses, and class-switch recombination. Following immune reconstitution of this novel strain with HSCs from a patient with immune dysregulation, polyendocrinopathy, enteropathy, X-linked (IPEX) syndrome, associated with aberrant FOXP3 function, mice developed a lethal inflammatory disorder with multiorgan involvement and autoantibody production mimicking the pathology seen in affected humans. This humanized mouse model permits in vivo evaluation of immune responses associated with genetically altered HSCs, including primary immunodeficiencies, and should facilitate the study of human immune pathobiology and the development of targeted therapeutics.
Collapse
|
572
|
Yamada E, Yoshikawa R, Nakano Y, Misawa N, Koyanagi Y, Sato K. Impacts of humanized mouse models on the investigation of HIV-1 infection: illuminating the roles of viral accessory proteins in vivo. Viruses 2015; 7:1373-90. [PMID: 25807049 PMCID: PMC4379576 DOI: 10.3390/v7031373] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Revised: 03/10/2015] [Accepted: 03/10/2015] [Indexed: 12/26/2022] Open
Abstract
Human immunodeficiency virus type 1 (HIV-1) encodes four accessory genes: vif, vpu, vpr, and nef. Recent investigations using in vitro cell culture systems have shed light on the roles of these HIV-1 accessory proteins, Vif, Vpr, Vpu, and Nef, in counteracting, modulating, and evading various cellular factors that are responsible for anti-HIV-1 intrinsic immunity. However, since humans are the exclusive target for HIV-1 infection, conventional animal models are incapable of mimicking the dynamics of HIV-1 infection in vivo. Moreover, the effects of HIV-1 accessory proteins on viral infection in vivo remain unclear. To elucidate the roles of HIV-1 accessory proteins in the dynamics of viral infection in vivo, humanized mouse models, in which the mice are xenotransplanted with human hematopoietic stem cells, has been utilized. This review describes the current knowledge of the roles of HIV-1 accessory proteins in viral infection, replication, and pathogenicity in vivo, which are revealed by the studies using humanized mouse models.
Collapse
Affiliation(s)
- Eri Yamada
- Laboratory of Viral Pathogenesis, Institute for Virus Research, Kyoto University, Kyoto 6068507, Japan.
| | - Rokusuke Yoshikawa
- Laboratory of Viral Pathogenesis, Institute for Virus Research, Kyoto University, Kyoto 6068507, Japan.
| | - Yusuke Nakano
- Laboratory of Viral Pathogenesis, Institute for Virus Research, Kyoto University, Kyoto 6068507, Japan.
| | - Naoko Misawa
- Laboratory of Viral Pathogenesis, Institute for Virus Research, Kyoto University, Kyoto 6068507, Japan.
| | - Yoshio Koyanagi
- Laboratory of Viral Pathogenesis, Institute for Virus Research, Kyoto University, Kyoto 6068507, Japan.
| | - Kei Sato
- Laboratory of Viral Pathogenesis, Institute for Virus Research, Kyoto University, Kyoto 6068507, Japan.
- CREST, Japan Science and Technology Agency, Saitama 3220012, Japan.
| |
Collapse
|
573
|
Lodhia KA, Hadley AM, Haluska P, Scott CL. Prioritizing therapeutic targets using patient-derived xenograft models. Biochim Biophys Acta Rev Cancer 2015; 1855:223-34. [PMID: 25783201 DOI: 10.1016/j.bbcan.2015.03.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Revised: 02/12/2015] [Accepted: 03/09/2015] [Indexed: 01/03/2023]
Abstract
Effective systemic treatment of cancer relies on the delivery of agents with optimal therapeutic potential. The molecular age of medicine has provided genomic tools that can identify a large number of potential therapeutic targets in individual patients, heralding the promise of personalized treatment. However, determining which potential targets actually drive tumor growth and should be prioritized for therapy is challenging. Indeed, reliable molecular matches of target and therapeutic agent have been stringently validated in the clinic for only a small number of targets. Patient-derived xenografts (PDXs) are tumor models developed in immunocompromised mice using tumor procured directly from the patient. As patient surrogates, PDX models represent a powerful tool for addressing individualized therapy. Challenges include humanizing the immune system of PDX models and ensuring high quality molecular annotation, in order to maximize insights for the clinic. Importantly, PDX can be sampled repeatedly and in parallel, to reveal clonal evolution, which may predict mechanisms of drug resistance and inform therapeutic strategy design.
Collapse
Affiliation(s)
- K A Lodhia
- Department of Oncology, Mayo Clinic, Rochester, MN, USA
| | - A M Hadley
- Stem Cells and Cancer Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
| | - P Haluska
- Department of Oncology, Mayo Clinic, Rochester, MN, USA
| | - C L Scott
- Stem Cells and Cancer Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia.
| |
Collapse
|
574
|
New humanized mouse model of bronchiolitis obliterans syndrome. Transplantation 2015; 99:468-9. [PMID: 25695785 DOI: 10.1097/tp.0000000000000631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Humanized animals are transplanted with human tissues and cells to study their behavior as they do in the human body. This commentary briefly summarizes the recent developments and discusses the limitations of these humanized animal models.
Collapse
|
575
|
Wermuth PJ, Jimenez SA. The significance of macrophage polarization subtypes for animal models of tissue fibrosis and human fibrotic diseases. Clin Transl Med 2015; 4:2. [PMID: 25852818 PMCID: PMC4384891 DOI: 10.1186/s40169-015-0047-4] [Citation(s) in RCA: 105] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Accepted: 01/20/2015] [Indexed: 12/15/2022] Open
Abstract
The systemic and organ-specific human fibrotic disorders collectively represent one of the most serious health problems world-wide causing a large proportion of the total world population mortality. The molecular pathways involved in their pathogenesis are complex and despite intensive investigations have not been fully elucidated. Whereas chronic inflammatory cell infiltration is universally present in fibrotic lesions, the central role of monocytes and macrophages as regulators of inflammation and fibrosis has only recently become apparent. However, the precise mechanisms involved in the contribution of monocytes/macrophages to the initiation, establishment, or progression of the fibrotic process remain largely unknown. Several monocyte and macrophage subpopulations have been identified, with certain phenotypes promoting inflammation whereas others display profibrotic effects. Given the unmet need for effective treatments for fibroproliferative diseases and the crucial regulatory role of monocyte/macrophage subpopulations in fibrogenesis, the development of therapeutic strategies that target specific monocyte/macrophage subpopulations has become increasingly attractive. We will provide here an overview of the current understanding of the role of monocyte/macrophage phenotype subpopulations in animal models of tissue fibrosis and in various systemic and organ-specific human fibrotic diseases. Furthermore, we will discuss recent approaches to the design of effective anti-fibrotic therapeutic interventions by targeting the phenotypic differences identified between the various monocyte and macrophage subpopulations.
Collapse
Affiliation(s)
- Peter J Wermuth
- Jefferson Institute of Molecular Medicine, Thomas Jefferson University, Bluemle Life Science Building Suite 509, 233 South 10th Street, Philadelphia, PA 19107-5541 USA
| | - Sergio A Jimenez
- Jefferson Institute of Molecular Medicine, Thomas Jefferson University, Bluemle Life Science Building Suite 509, 233 South 10th Street, Philadelphia, PA 19107-5541 USA
| |
Collapse
|
576
|
Abstract
The complexity and heterogeneity of ovarian cancer cases are difficult to reproduce in in vitro studies, which cannot adequately elucidate the molecular events involved in tumor initiation and disease metastasis. It has now become clear that, although the multiple histological subtypes of ovarian cancer are being treated with similar surgical and therapeutic approaches, they are in fact characterized by distinct phenotypes, cell of origin, and underlying key genetic and genomic alterations. Consequently, the development of more personalized treatment methodologies, which are aimed at improving patient care and prognosis, will greatly benefit from a better understanding of the key differences between various subtypes. To accomplish this, animal models of all histotypes need to be generated in order to provide accurate in vivo platforms for research and the testing of targeted treatments and immune therapies. Both genetically engineered mouse models (GEMMs) and xenograft models have the ability to further our understanding of key mechanisms facilitating tumorigenesis, and at the same time offer insight into enhanced imaging and treatment modalities. While genetic models may be better suited to examine oncogenic functions and interactions during tumorigenesis, patient-derived xenografts (PDXs) are likely a superior model to assess drug efficacy, especially in concurrent clinical trials, due to their similarity to the tumors from which they are derived. Genetic and avatar models possess great clinical utility and have both benefits and limitations. Additionally, the laying hen model, which spontaneously develops ovarian tumors, has inherent advantages for the study of epithelial ovarian cancer (EOC) and recent work champions this model especially when assessing chemoprevention strategies. While high-grade ovarian serous tumors are the most prevalent form of EOC, rarer ovarian cancer variants, such as small cell ovarian carcinoma of the hypercalcemic type and transitional cell carcinoma, or non-epithelial tumors, including germ cell tumors, will also benefit from the generation of improved models to advance our understanding of tumorigenic mechanisms and the development of selective therapeutic options.
Collapse
Affiliation(s)
- Noor Hasan
- Department of Pathology, Division of Women's and Perinatal Pathology, Eugene Braunwald Research Center, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Anders W Ohman
- Department of Pathology, Division of Women's and Perinatal Pathology, Eugene Braunwald Research Center, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Daniela M Dinulescu
- Department of Pathology, Division of Women's and Perinatal Pathology, Eugene Braunwald Research Center, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
577
|
Riccardo F, Aurisicchio L, Impellizeri JA, Cavallo F. The importance of comparative oncology in translational medicine. Cancer Immunol Immunother 2015; 64:137-48. [PMID: 25548094 PMCID: PMC11029667 DOI: 10.1007/s00262-014-1645-5] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Accepted: 12/15/2014] [Indexed: 12/20/2022]
Abstract
Human cancer is so complex that in vivo preclinical models are needed if effective therapies are to be developed. Naturally occurring cancers in companion animals are therefore a great resource, as shown by the remarkable growth that comparative oncology has seen over the last 30 years. Cancer has become a leading cause of death in companion animals now that more pets are living long enough to develop the disease. Furthermore, more owners are seeking advanced and novel therapies for their pets as they are very much considered family members. Living in the same environments, pets and humans are often afflicted by the same types of cancer which show similar behavior and, in some species, express the same antigen molecules. The treatment of pet tumors using novel therapies is of compelling translational significance.
Collapse
Affiliation(s)
- Federica Riccardo
- Department of Molecular Biotechnologies and Health Sciences, Molecular Biotechnology Center, University of Turin, Via Nizza, 52, 10126 Turin, Italy
| | | | | | - Federica Cavallo
- Department of Molecular Biotechnologies and Health Sciences, Molecular Biotechnology Center, University of Turin, Via Nizza, 52, 10126 Turin, Italy
| |
Collapse
|
578
|
McIntosh BE, Brown ME, Duffin BM, Maufort JP, Vereide DT, Slukvin II, Thomson JA. Nonirradiated NOD,B6.SCID Il2rγ-/- Kit(W41/W41) (NBSGW) mice support multilineage engraftment of human hematopoietic cells. Stem Cell Reports 2015; 4:171-80. [PMID: 25601207 PMCID: PMC4325197 DOI: 10.1016/j.stemcr.2014.12.005] [Citation(s) in RCA: 169] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Revised: 12/06/2014] [Accepted: 12/08/2014] [Indexed: 12/22/2022] Open
Abstract
In this study, we demonstrate a newly derived mouse model that supports engraftment of human hematopoietic stem cells (HSCs) in the absence of irradiation. We cross the NOD.Cg-PrkdcscidIl2rgtm1Wjl/SzJ (NSG) strain with the C57BL/6J-KitW-41J/J (C57BL/6.KitW41) strain and engraft, without irradiation, the resulting NBSGW strain with human cord blood CD34+ cells. At 12-weeks postengraftment in NBSGW mice, we observe human cell chimerism in marrow (97% ± 0.4%), peripheral blood (61% ± 2%), and spleen (94% ± 2%) at levels observed with irradiation in NSG mice. We also detected a significant number of glycophorin-A-positive expressing cells in the developing NBSGW marrow. Further, the observed levels of human hematopoietic chimerism mimic those reported for both irradiated NSG and NSG-transgenic strains. This mouse model permits HSC engraftment while avoiding the complicating hematopoietic, gastrointestinal, and neurological side effects associated with irradiation and allows investigators without access to radiation to pursue engraftment studies with human HSCs. In engraftment experiments, nonirradiated NBSGW mice show enhanced humanization Similar levels of human chimerism are observed between both irNSG and NBSGW mice NBSGW mice are conducive to serial transplantation without irradiation NBSGW mice harbor a mutant KitW41 allele, aiding Gly-A+ development in the marrow
Collapse
Affiliation(s)
| | - Matthew E Brown
- Department of Surgery, University of Wisconsin, Madison, WI 53715, USA
| | - Bret M Duffin
- Morgridge Institute for Research, Madison, WI 53715, USA
| | - John P Maufort
- Morgridge Institute for Research, Madison, WI 53715, USA
| | - David T Vereide
- Morgridge Institute for Research, Madison, WI 53715, USA; Biotechnology Center, University of Wisconsin, Madison, WI 53706, USA
| | - Igor I Slukvin
- Wisconsin National Primate Research Center, University of Wisconsin, Madison, WI 53715, USA; Department of Pathology and Laboratory Medicine, University of Wisconsin, Madison, WI 53715, USA
| | - James A Thomson
- Morgridge Institute for Research, Madison, WI 53715, USA; Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53706, USA; Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, Santa Barbara, CA 93106, USA.
| |
Collapse
|
579
|
Broad CTL response is required to clear latent HIV-1 due to dominance of escape mutations. Nature 2015; 517:381-5. [PMID: 25561180 PMCID: PMC4406054 DOI: 10.1038/nature14053] [Citation(s) in RCA: 427] [Impact Index Per Article: 42.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Accepted: 11/11/2014] [Indexed: 02/05/2023]
Abstract
Despite antiretroviral therapy (ART), human immunodeficiency virus (HIV)-1 persists in a stable latent reservoir, primarily in resting memory CD4(+) T cells. This reservoir presents a major barrier to the cure of HIV-1 infection. To purge the reservoir, pharmacological reactivation of latent HIV-1 has been proposed and tested both in vitro and in vivo. A key remaining question is whether virus-specific immune mechanisms, including cytotoxic T lymphocytes (CTLs), can clear infected cells in ART-treated patients after latency is reversed. Here we show that there is a striking all or none pattern for CTL escape mutations in HIV-1 Gag epitopes. Unless ART is started early, the vast majority (>98%) of latent viruses carry CTL escape mutations that render infected cells insensitive to CTLs directed at common epitopes. To solve this problem, we identified CTLs that could recognize epitopes from latent HIV-1 that were unmutated in every chronically infected patient tested. Upon stimulation, these CTLs eliminated target cells infected with autologous virus derived from the latent reservoir, both in vitro and in patient-derived humanized mice. The predominance of CTL-resistant viruses in the latent reservoir poses a major challenge to viral eradication. Our results demonstrate that chronically infected patients retain a broad-spectrum viral-specific CTL response and that appropriate boosting of this response may be required for the elimination of the latent reservoir.
Collapse
|
580
|
Takahashi T, Katano I, Ito R, Ito M. Visualization of the human CD4⁺ T-cell response in humanized HLA-DR4-expressing NOD/Shi-scid/γc(null) (NOG) mice by retrogenic expression of the human TCR gene. Biochem Biophys Res Commun 2015; 456:219-24. [PMID: 25462565 DOI: 10.1016/j.bbrc.2014.11.062] [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: 11/10/2014] [Accepted: 11/13/2014] [Indexed: 10/24/2022]
Abstract
The development of severe immunodeficient mouse strains containing various human genes, including cytokines or HLA, has enabled the reconstitution of functional human immune systems after transplantation of human hematopoietic stem cells (HSC). Accumulating evidence has suggested that HLA-restricted antigen-specific human T-cell responses can be generated in these humanized mice. To directly monitor immune responses of human CD4(+) T cells, we introduced β-lactoglobulin (BLG)-specific T cell receptor (TCR) genes derived from CD4(+) T-cell clones of cow-milk allergy patients into HSCs, and subsequently transplanted them into NOG-HLA-DR4 transgenic/I-Aβ deficient mice (NOG-DR4/I-A(o)). In the thymus, thymocytes with BLG-specific TCR preferentially differentiated into CD4(+)CD8(-) single-positive cells. Adoptive transfer of mature CD4(+) T cells expressing the TCR into recipient NOG-DR4/I-A(o) mice demonstrated that human CD4(+) T cells proliferated in response to antigenic stimulation and produced IFN-γ in vivo, suggesting that functional T-cell reactions (especially Th1-skewed responses) were induced in humanized mice.
Collapse
Affiliation(s)
- Takeshi Takahashi
- Central Institute for Experimental Animals, 3-25-12 Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan.
| | - Ikumi Katano
- Central Institute for Experimental Animals, 3-25-12 Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
| | - Ryoji Ito
- Central Institute for Experimental Animals, 3-25-12 Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
| | - Mamoru Ito
- Central Institute for Experimental Animals, 3-25-12 Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
| |
Collapse
|
581
|
Li Y, Di Santo JP. Probing Human NK Cell Biology Using Human Immune System (HIS) Mice. Curr Top Microbiol Immunol 2015; 395:191-208. [PMID: 26459320 DOI: 10.1007/82_2015_488] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Our incomplete understanding of the mechanisms that orchestrate human lymphocyte differentiation and condition human immune responses is in part due to the limited access to normal human tissue samples that can inform on these complex processes. In addition, in vitro culture conditions fail to recapitulate the three-dimensional microenvironments that influence cell-cell interactions and impact on immune outcomes. Small animals provide a preclinical model to dissect and probe immunity and over the past decades, development of immunodeficient hosts that can be engrafted with human hematopoietic precursors and mature cells have led to the development of new in vivo models to study human lymphocyte development and function. Natural killer (NK) cells are implicated in the recognition and elimination of pathogen-infected and transformed cells and belong to a family of diverse innate lymphoid cells (ILCs) that provide early immune defense against disease. Here, we summarize the use of humanized mouse models for the study of NK cell and group 1 ILCs and their respective roles in immunity and tissue homeostasis.
Collapse
Affiliation(s)
- Yan Li
- Innate Immunity Unit, Institut Pasteur, 25 rue du Docteur Roux, Paris, 75724, France.,Inserm U668, Paris, France
| | - James P Di Santo
- Innate Immunity Unit, Institut Pasteur, 25 rue du Docteur Roux, Paris, 75724, France. .,Inserm U668, Paris, France.
| |
Collapse
|
582
|
Abstract
Breast cancer is the most common cause of cancer death in women worldwide. This malignancy is a complex disease, which is defined by an intrinsic heterogeneity on the histopathological and molecular level as well as response to therapy and outcome. In addition to classical histopathological features, breast cancer can be categorized into at least five major subtypes based on comprehensive gene expression profiling: luminal A, luminal B, basal-like, ERBB2-positive, and normal-like breast cancer. Genetically engineered mouse models can serve as tools to study the molecular underpinnings for this disease. Given the genetic complexity that drives the initiation and progression of individual breast cancer subtypes, it is evident that certain models can reflect only particular aspects of this malignancy. In this book chapter, we will primarily focus on advances in modeling breast cancer at defined stages of carcinogenesis using genetically engineered mice. We will discuss the ability as well as shortcomings of these models to faithfully recapitulate the spectrum of human breast cancer subtypes.
Collapse
|
583
|
Klemm F, Joyce JA. Microenvironmental regulation of therapeutic response in cancer. Trends Cell Biol 2014; 25:198-213. [PMID: 25540894 DOI: 10.1016/j.tcb.2014.11.006] [Citation(s) in RCA: 545] [Impact Index Per Article: 49.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Revised: 11/20/2014] [Accepted: 11/21/2014] [Indexed: 02/08/2023]
Abstract
The tumor microenvironment (TME) not only plays a pivotal role during cancer progression and metastasis but also has profound effects on therapeutic efficacy. In the case of microenvironment-mediated resistance this can involve an intrinsic response, including the co-option of pre-existing structural elements and signaling networks, or an acquired response of the tumor stroma following the therapeutic insult. Alternatively, in other contexts, the TME has a multifaceted ability to enhance therapeutic efficacy. This review examines recent advances in our understanding of the contribution of the TME during cancer therapy and discusses key concepts that may be amenable to therapeutic intervention.
Collapse
Affiliation(s)
- Florian Klemm
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Johanna A Joyce
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| |
Collapse
|
584
|
Choi SYC, Lin D, Gout PW, Collins CC, Xu Y, Wang Y. Lessons from patient-derived xenografts for better in vitro modeling of human cancer. Adv Drug Deliv Rev 2014; 79-80:222-37. [PMID: 25305336 DOI: 10.1016/j.addr.2014.09.009] [Citation(s) in RCA: 126] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2013] [Revised: 09/02/2014] [Accepted: 09/23/2014] [Indexed: 12/21/2022]
Abstract
The development of novel cancer therapeutics is often plagued by discrepancies between drug efficacies obtained in preclinical studies and outcomes of clinical trials. The inconsistencies can be attributed to a lack of clinical relevance of the cancer models used for drug testing. While commonly used in vitro culture systems are advantageous for addressing specific experimental questions, they are often gross, fidelity-lacking simplifications that largely ignore the heterogeneity of cancers as well as the complexity of the tumor microenvironment. Factors such as tumor architecture, interactions among cancer cells and between cancer and stromal cells, and an acidic tumor microenvironment are critical characteristics observed in patient-derived cancer xenograft models and in the clinic. By mimicking these crucial in vivo characteristics through use of 3D cultures, co-culture systems and acidic culture conditions, an in vitro cancer model/microenvironment that is more physiologically relevant may be engineered to produce results more readily applicable to the clinic.
Collapse
Affiliation(s)
- Stephen Yiu Chuen Choi
- Department of Experimental Therapeutics, BC Cancer Agency, Vancouver, BC, Canada; Vancouver Prostate Centre, Vancouver, BC, Canada.
| | - Dong Lin
- Department of Experimental Therapeutics, BC Cancer Agency, Vancouver, BC, Canada; Vancouver Prostate Centre, Vancouver, BC, Canada.
| | - Peter W Gout
- Department of Experimental Therapeutics, BC Cancer Agency, Vancouver, BC, Canada.
| | - Colin C Collins
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada; Vancouver Prostate Centre, Vancouver, BC, Canada.
| | - Yong Xu
- Department of Urology, Second Affiliated Hospital of Tianjin Medical University, Tianjin, P.R. China.
| | - Yuzhuo Wang
- Department of Experimental Therapeutics, BC Cancer Agency, Vancouver, BC, Canada; Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada; Vancouver Prostate Centre, Vancouver, BC, Canada.
| |
Collapse
|
585
|
Ivanisevic J, Epstein A, Kurczy ME, Benton HP, Uritboonthai W, Fox HS, Boska MD, Gendelman HE, Siuzdak G. Brain region mapping using global metabolomics. CHEMISTRY & BIOLOGY 2014; 21:1575-84. [PMID: 25457182 PMCID: PMC4304924 DOI: 10.1016/j.chembiol.2014.09.016] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Revised: 09/05/2014] [Accepted: 09/18/2014] [Indexed: 11/17/2022]
Abstract
Historically, studies of brain metabolism have been based on targeted analyses of a limited number of metabolites. Here we present an untargeted mass spectrometry-based metabolomic strategy that has successfully uncovered differences in a broad array of metabolites across anatomical regions of the mouse brain. The NSG immunodeficient mouse model was chosen because of its ability to undergo humanization leading to numerous applications in oncology and infectious disease research. Metabolic phenotyping by hydrophilic interaction liquid chromatography and nanostructure imaging mass spectrometry revealed both water-soluble and lipid metabolite patterns across brain regions. Neurochemical differences in metabolic phenotypes were mainly defined by various phospholipids and several intriguing metabolites including carnosine, cholesterol sulfate, lipoamino acids, uric acid, and sialic acid, whose physiological roles in brain metabolism are poorly understood. This study helps define regional homeostasis for the normal mouse brain to give context to the reaction to pathological events.
Collapse
Affiliation(s)
- Julijana Ivanisevic
- Scripps Center for Metabolomics and Mass Spectrometry, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037
| | - Adrian Epstein
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5880
| | - Michael E. Kurczy
- Scripps Center for Metabolomics and Mass Spectrometry, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037
| | - H. Paul Benton
- Scripps Center for Metabolomics and Mass Spectrometry, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037
| | - Winnie Uritboonthai
- Scripps Center for Metabolomics and Mass Spectrometry, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037
| | - Howard S. Fox
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5880
| | - Michael D. Boska
- Department of Radiology, University of Nebraska Medical Center, Omaha, NE 68198-5880
| | - Howard E. Gendelman
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5880
| | - Gary Siuzdak
- Scripps Center for Metabolomics and Mass Spectrometry, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037
| |
Collapse
|
586
|
Epigenetic and in vivo comparison of diverse MSC sources reveals an endochondral signature for human hematopoietic niche formation. Blood 2014; 125:249-60. [PMID: 25406351 DOI: 10.1182/blood-2014-04-572255] [Citation(s) in RCA: 176] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
In the last decade there has been a rapid expansion in clinical trials using mesenchymal stromal cells (MSCs) from a variety of tissues. However, despite similarities in morphology, immunophenotype, and differentiation behavior in vitro, MSCs sourced from distinct tissues do not necessarily have equivalent biological properties. We performed a genome-wide methylation, transcription, and in vivo evaluation of MSCs from human bone marrow (BM), white adipose tissue, umbilical cord, and skin cultured in humanized media. Surprisingly, only BM-derived MSCs spontaneously formed a BM cavity through a vascularized cartilage intermediate in vivo that was progressively replaced by hematopoietic tissue and bone. Only BM-derived MSCs exhibited a chondrogenic transcriptional program with hypomethylation and increased expression of RUNX3, RUNX2, BGLAP, MMP13, and ITGA10 consistent with a latent and primed skeletal developmental potential. The humanized MSC-derived microenvironment permitted homing and maintenance of long-term murine SLAM(+) hematopoietic stem cells (HSCs), as well as human CD34(+)/CD38(-)/CD90(+)/CD45RA(+) HSCs after cord blood transplantation. These studies underscore the profound differences in developmental potential between MSC sources independent of donor age, with implications for their clinical use. We also demonstrate a tractable human niche model for studying homing and engraftment of human hematopoietic cells in normal and neoplastic states.
Collapse
|
587
|
Weill JC, Reynaud CA. [Ever more humanized mice for new therapeutic applications]. Med Sci (Paris) 2014; 30:949-51. [PMID: 25388574 DOI: 10.1051/medsci/20143011006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Affiliation(s)
- Jean-Claude Weill
- Institut Necker-Enfants malades (INEM), Inserm U1151 - CNRS UMR 8253, université Paris Descartes, Sorbonne Paris Cité, faculté de médecine-site Broussais, 14, rue Maria Helena Viera Da Silva, 75993 Paris Cedex 14, France
| | - Claude-Agnès Reynaud
- Institut Necker-Enfants malades (INEM), Inserm U1151 - CNRS UMR 8253, université Paris Descartes, Sorbonne Paris Cité, faculté de médecine-site Broussais, 14, rue Maria Helena Viera Da Silva, 75993 Paris Cedex 14, France
| |
Collapse
|
588
|
Kepp O, Senovilla L, Vitale I, Vacchelli E, Adjemian S, Agostinis P, Apetoh L, Aranda F, Barnaba V, Bloy N, Bracci L, Breckpot K, Brough D, Buqué A, Castro MG, Cirone M, Colombo MI, Cremer I, Demaria S, Dini L, Eliopoulos AG, Faggioni A, Formenti SC, Fučíková J, Gabriele L, Gaipl US, Galon J, Garg A, Ghiringhelli F, Giese NA, Guo ZS, Hemminki A, Herrmann M, Hodge JW, Holdenrieder S, Honeychurch J, Hu HM, Huang X, Illidge TM, Kono K, Korbelik M, Krysko DV, Loi S, Lowenstein PR, Lugli E, Ma Y, Madeo F, Manfredi AA, Martins I, Mavilio D, Menger L, Merendino N, Michaud M, Mignot G, Mossman KL, Multhoff G, Oehler R, Palombo F, Panaretakis T, Pol J, Proietti E, Ricci JE, Riganti C, Rovere-Querini P, Rubartelli A, Sistigu A, Smyth MJ, Sonnemann J, Spisek R, Stagg J, Sukkurwala AQ, Tartour E, Thorburn A, Thorne SH, Vandenabeele P, Velotti F, Workenhe ST, Yang H, Zong WX, Zitvogel L, Kroemer G, Galluzzi L. Consensus guidelines for the detection of immunogenic cell death. Oncoimmunology 2014; 3:e955691. [PMID: 25941621 PMCID: PMC4292729 DOI: 10.4161/21624011.2014.955691] [Citation(s) in RCA: 640] [Impact Index Per Article: 58.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Accepted: 08/04/2014] [Indexed: 02/07/2023] Open
Abstract
Apoptotic cells have long been considered as intrinsically tolerogenic or unable to elicit immune responses specific for dead cell-associated antigens. However, multiple stimuli can trigger a functionally peculiar type of apoptotic demise that does not go unnoticed by the adaptive arm of the immune system, which we named "immunogenic cell death" (ICD). ICD is preceded or accompanied by the emission of a series of immunostimulatory damage-associated molecular patterns (DAMPs) in a precise spatiotemporal configuration. Several anticancer agents that have been successfully employed in the clinic for decades, including various chemotherapeutics and radiotherapy, can elicit ICD. Moreover, defects in the components that underlie the capacity of the immune system to perceive cell death as immunogenic negatively influence disease outcome among cancer patients treated with ICD inducers. Thus, ICD has profound clinical and therapeutic implications. Unfortunately, the gold-standard approach to detect ICD relies on vaccination experiments involving immunocompetent murine models and syngeneic cancer cells, an approach that is incompatible with large screening campaigns. Here, we outline strategies conceived to detect surrogate markers of ICD in vitro and to screen large chemical libraries for putative ICD inducers, based on a high-content, high-throughput platform that we recently developed. Such a platform allows for the detection of multiple DAMPs, like cell surface-exposed calreticulin, extracellular ATP and high mobility group box 1 (HMGB1), and/or the processes that underlie their emission, such as endoplasmic reticulum stress, autophagy and necrotic plasma membrane permeabilization. We surmise that this technology will facilitate the development of next-generation anticancer regimens, which kill malignant cells and simultaneously convert them into a cancer-specific therapeutic vaccine.
Collapse
Key Words
- APC, antigen-presenting cell
- ATF6, activating transcription factor 6
- ATP release
- BAK1, BCL2-antagonist/killer 1
- BAX, BCL2-associated X protein
- BCL2, B-cell CLL/lymphoma 2 protein
- CALR, calreticulin
- CTL, cytotoxic T lymphocyte
- DAMP, damage-associated molecular pattern
- DAPI, 4′,6-diamidino-2-phenylindole
- DiOC6(3), 3,3′-dihexyloxacarbocyanine iodide
- EIF2A, eukaryotic translation initiation factor 2A
- ER, endoplasmic reticulum
- FLT3LG, fms-related tyrosine kinase 3 ligand
- G3BP1, GTPase activating protein (SH3 domain) binding protein 1
- GFP, green fluorescent protein
- H2B, histone 2B
- HMGB1
- HMGB1, high mobility group box 1
- HSP, heat shock protein
- HSV-1, herpes simplex virus type I
- ICD, immunogenic cell death
- IFN, interferon
- IL, interleukin
- MOMP, mitochondrial outer membrane permeabilization
- PDIA3, protein disulfide isomerase family A
- PI, propidium iodide
- RFP, red fluorescent protein
- TLR, Toll-like receptor
- XBP1, X-box binding protein 1
- autophagy
- calreticulin
- endoplasmic reticulum stress
- immunotherapy
- member 3
- Δψm, mitochondrial transmembrane potential
Collapse
Affiliation(s)
- Oliver Kepp
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Center de Recherche des Cordeliers; Paris, France
- INSERM; U1138; Paris, France
- Metabolomics and Cell Biology Platforms; Gustave Roussy Cancer Campus; Villejuif, France
| | - Laura Senovilla
- INSERM; U1138; Paris, France
- Metabolomics and Cell Biology Platforms; Gustave Roussy Cancer Campus; Villejuif, France
- INSERM; U1015; Villejuif, France
| | - Ilio Vitale
- Regina Elena National Cancer Institute; Rome, Italy
| | - Erika Vacchelli
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Center de Recherche des Cordeliers; Paris, France
- INSERM; U1138; Paris, France
- Gustave Roussy Cancer Campus; Villejuif, France
| | - Sandy Adjemian
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Center de Recherche des Cordeliers; Paris, France
- INSERM; U1138; Paris, France
- Gustave Roussy Cancer Campus; Villejuif, France
- Molecular Cell Biology Laboratory; Department of Immunology; Institute of Biomedical Sciences; University of São Paulo; São Paulo, Brazil
| | - Patrizia Agostinis
- Cell Death Research and Therapy (CDRT) Laboratory; Department of Cellular and Molecular Medicine; University of Leuven; Leuven, Belgium
| | - Lionel Apetoh
- INSERM; UMR866; Dijon, France
- Centre Georges François Leclerc; Dijon, France
- Université de Bourgogne; Dijon, France
| | - Fernando Aranda
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Center de Recherche des Cordeliers; Paris, France
- INSERM; U1138; Paris, France
- Gustave Roussy Cancer Campus; Villejuif, France
| | - Vincenzo Barnaba
- Departement of Internal Medicine and Medical Sciences; University of Rome La Sapienza; Rome, Italy
- Istituto Pasteur; Fondazione Cenci Bolognetti; Rome, Italy
| | - Norma Bloy
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Center de Recherche des Cordeliers; Paris, France
- INSERM; U1138; Paris, France
- Gustave Roussy Cancer Campus; Villejuif, France
| | - Laura Bracci
- Department of Hematology; Oncology and Molecular Medicine; Istituto Superiore di Sanità (ISS); Rome, Italy
| | - Karine Breckpot
- Laboratory of Molecular and Cellular Therapy (LMCT); Department of Biomedical Sciences Medical School of the Free University of Brussels (VUB); Jette, Belgium
| | - David Brough
- Faculty of Life Sciences; University of Manchester; Manchester, UK
| | - Aitziber Buqué
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Center de Recherche des Cordeliers; Paris, France
- INSERM; U1138; Paris, France
- Gustave Roussy Cancer Campus; Villejuif, France
| | - Maria G. Castro
- Department of Neurosurgery and Cell and Developmental Biology; University of Michigan School of Medicine; Ann Arbor, MI USA
| | - Mara Cirone
- Department of Experimental Medicine; University of Rome La Sapienza; Rome, Italy
| | - Maria I. Colombo
- Laboratorio de Biología Celular y Molecular; Instituto de Histología y Embriología (IHEM); Facultad de Ciencias Médicas; Universidad Nacional de Cuyo; CONICET; Mendoza, Argentina
| | - Isabelle Cremer
- INSERM; U1138; Paris, France
- Université Pierre et Marie Curie/Paris VI; Paris, France
- Equipe 13; Center de Recherche des Cordeliers; Paris, France
| | - Sandra Demaria
- Department of Pathology; New York University School of Medicine; New York, NY USA
| | - Luciana Dini
- Department of Biological and Environmental Science and Technology (DiSTeBA); University of Salento; Lecce, Italy
| | - Aristides G. Eliopoulos
- Molecular and Cellular Biology Laboratory; Division of Basic Sciences; University of Crete Medical School; Heraklion, Greece
- Institute of Molecular Biology and Biotechnology; Foundation of Research and Technology - Hellas; Heraklion, Greece
| | - Alberto Faggioni
- Department of Experimental Medicine; University of Rome La Sapienza; Rome, Italy
| | - Silvia C. Formenti
- Department of Radiation Oncology; NewYork University School of Medicine and Langone Medical Center; New York, NY USA
| | - Jitka Fučíková
- Department of Immunology; 2 Faculty of Medicine and University Hospital Motol, Charles University; Prague, Czech Republic
- Sotio; Prague, Czech Republic
| | - Lucia Gabriele
- Department of Hematology; Oncology and Molecular Medicine; Istituto Superiore di Sanità (ISS); Rome, Italy
| | - Udo S. Gaipl
- Department of Radiation Oncology; University Hospital Erlangen; University of Erlangen-Nürnberg; Erlangen, Germany
| | - Jérôme Galon
- INSERM; U1138; Paris, France
- Université Pierre et Marie Curie/Paris VI; Paris, France
- Université Paris Descartes/Paris V; Sorbonne Paris Cité; Paris, France
- Laboratory of Integrative Cancer Immunology; Center de Recherche des Cordeliers; Paris, France
| | - Abhishek Garg
- Cell Death Research and Therapy (CDRT) Laboratory; Department of Cellular and Molecular Medicine; University of Leuven; Leuven, Belgium
| | - François Ghiringhelli
- INSERM; UMR866; Dijon, France
- Centre Georges François Leclerc; Dijon, France
- Université de Bourgogne; Dijon, France
| | - Nathalia A. Giese
- European Pancreas Center; Department of Surgery; University Hospital Heidelberg; Heidelberg, Germany
| | - Zong Sheng Guo
- Department of Surgery; University of Pittsburgh; Pittsburgh, PA USA
| | - Akseli Hemminki
- Cancer Gene Therapy Group; Transplantation laboratory; Haartman Institute; University of Helsinki; Helsinki, Finland
| | - Martin Herrmann
- Department of Internal Medicine 3; University of Erlangen-Nuremberg; Erlangen, Germany
| | - James W. Hodge
- Laboratory of Tumor Immunology and Biology; Center for Cancer Research; National Cancer Institute (NCI), National Institutes of Health (NIH); Bethesda, MD USA
| | - Stefan Holdenrieder
- Institute of Clinical Chemistry and Clinical Pharmacology; University Hospital Bonn; Bonn, Germany
| | - Jamie Honeychurch
- Faculty of Medical and Human Sciences, Institute of Cancer Studies; Manchester Academic Health Sciences Center; University of Manchester; Manchester, UK
| | - Hong-Min Hu
- Cancer Research and Biotherapy Center; Second Affiliated Hospital of Southeast University; Nanjing, China
- Laboratory of Cancer Immunobiology; Earle A. Chiles Research Institute; Providence Portland Medical Center; Portland, OR USA
| | - Xing Huang
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Center de Recherche des Cordeliers; Paris, France
- INSERM; U1138; Paris, France
- Metabolomics and Cell Biology Platforms; Gustave Roussy Cancer Campus; Villejuif, France
| | - Tim M. Illidge
- Faculty of Medical and Human Sciences, Institute of Cancer Studies; Manchester Academic Health Sciences Center; University of Manchester; Manchester, UK
| | - Koji Kono
- Department of Surgery; National University of Singapore; Singapore, Singapore
- Cancer Science Institute of Singapore; National University of Singapore; Singapore, Singapore
| | | | - Dmitri V. Krysko
- VIB Inflammation Research Center; Ghent, Belgium
- Department of Biomedical Molecular Biology; Ghent University; Ghent, Belgium
| | - Sherene Loi
- Division of Cancer Medicine and Division of Research; Peter MacCallum Cancer Center; East Melbourne; Victoria, Australia
| | - Pedro R. Lowenstein
- Department of Neurosurgery and Cell and Developmental Biology; University of Michigan School of Medicine; Ann Arbor, MI USA
| | - Enrico Lugli
- Unit of Clinical and Experimental Immunology; Humanitas Clinical and Research Center; Milan, Italy
- Department of Medical Biotechnologies and Translational Medicine, University of Milan; Rozzano, Italy
| | - Yuting Ma
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Center de Recherche des Cordeliers; Paris, France
- INSERM; U1138; Paris, France
- Gustave Roussy Cancer Campus; Villejuif, France
| | - Frank Madeo
- Institute of Molecular Biosciences; University of Graz; Graz, Austria
| | - Angelo A. Manfredi
- University Vita-Salute San Raffaele; Milano, Italy
- San Raffaele Scientific Institute; Milano, Italy
| | - Isabelle Martins
- Gustave Roussy Cancer Campus; Villejuif, France
- INSERM, U1030; Villejuif, France
- Faculté de Médecine; Université Paris-Sud/Paris XI; Kremlin-Bicêtre, France
| | - Domenico Mavilio
- Unit of Clinical and Experimental Immunology; Humanitas Clinical and Research Center; Milan, Italy
- Department of Medical Biotechnologies and Translational Medicine, University of Milan; Rozzano, Italy
| | - Laurie Menger
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Center de Recherche des Cordeliers; Paris, France
- INSERM; U1138; Paris, France
- Gustave Roussy Cancer Campus; Villejuif, France
- Cancer Immunology Unit, Research Department of Haematology; University College London (UCL) Cancer Institute; London, UK
| | - Nicolò Merendino
- Department of Ecological and Biological Sciences (DEB), Tuscia University; Viterbo, Italy
| | - Michael Michaud
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Center de Recherche des Cordeliers; Paris, France
- INSERM; U1138; Paris, France
- Gustave Roussy Cancer Campus; Villejuif, France
| | - Gregoire Mignot
- Cellular and Molecular Immunology and Endocrinology, Oniris; Nantes, France
| | - Karen L. Mossman
- Department of Pathology and Molecular Medicine; McMaster Immunology Research Center; Hamilton, Canada
- Institute for Infectious Disease Research; McMaster University; Hamilton, Canada
| | - Gabriele Multhoff
- Department of Radiation Oncology; Klinikum rechts der Isar; Technical University of Munich; Munich, Germany
| | - Rudolf Oehler
- Comprehensive Cancer Center; Medical University of Vienna; Vienna, Austria
| | - Fabio Palombo
- Departement of Internal Medicine and Medical Sciences; University of Rome La Sapienza; Rome, Italy
- Istituto Pasteur; Fondazione Cenci Bolognetti; Rome, Italy
| | | | - Jonathan Pol
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Center de Recherche des Cordeliers; Paris, France
- INSERM; U1138; Paris, France
- Gustave Roussy Cancer Campus; Villejuif, France
| | - Enrico Proietti
- Department of Hematology; Oncology and Molecular Medicine; Istituto Superiore di Sanità (ISS); Rome, Italy
| | - Jean-Ehrland Ricci
- INSERM; U1065; Nice, France
- Equipe “Contrôle Métabolique des Morts Cellulaires,” Center Méditerranéen de Médecine Moléculaire (C3M); Nice, France
- Faculté de Médecine; Université de Nice Sophia Antipolis; Nice, France
- Centre Hospitalier Universitaire de Nice; Nice, France
| | - Chiara Riganti
- Department of Oncology and Subalpine Center for Research and Experimental Medicine (CeRMS); University of Turin; Turin, Italy
| | - Patrizia Rovere-Querini
- University Vita-Salute San Raffaele; Milano, Italy
- San Raffaele Scientific Institute; Milano, Italy
| | - Anna Rubartelli
- Cell Biology Unit; Azienda Ospedaliera Universitaria San Martino; Istituto Nazionale per la Ricerca sul Cancro; Genova, Italy
| | | | - Mark J. Smyth
- Immunology in Cancer and Infection Laboratory; QIMR Berghofer Medical Research Institute; Herston, Australia
- School of Medicine, University of Queensland; Herston, Australia
| | - Juergen Sonnemann
- Department of Pediatric Haematology and Oncology; Jena University Hospital, Children's Clinic; Jena, Germany
| | - Radek Spisek
- Department of Immunology; 2 Faculty of Medicine and University Hospital Motol, Charles University; Prague, Czech Republic
- Sotio; Prague, Czech Republic
| | - John Stagg
- Centre de Recherche du Center Hospitalier de l’Université de Montréal; Faculté de Pharmacie, Université de Montréal; Montréal, Canada
| | - Abdul Qader Sukkurwala
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Center de Recherche des Cordeliers; Paris, France
- INSERM; U1138; Paris, France
- Gustave Roussy Cancer Campus; Villejuif, France
- Department of Pathology, Dow International Medical College; Dow University of Health Sciences; Karachi, Pakistan
| | - Eric Tartour
- INSERM; U970; Paris, France
- Pôle de Biologie; Hôpital Européen Georges Pompidou; AP-HP; Paris, France
| | - Andrew Thorburn
- Department of Pharmacology; University of Colorado School of Medicine; Aurora, CO USA
| | | | - Peter Vandenabeele
- VIB Inflammation Research Center; Ghent, Belgium
- Department of Biomedical Molecular Biology; Ghent University; Ghent, Belgium
- Methusalem Program; Ghent University; Ghent, Belgium
| | - Francesca Velotti
- Department of Ecological and Biological Sciences (DEB), Tuscia University; Viterbo, Italy
| | - Samuel T. Workenhe
- Department of Pathology and Molecular Medicine; McMaster Immunology Research Center; Hamilton, Canada
- Institute for Infectious Disease Research; McMaster University; Hamilton, Canada
| | - Haining Yang
- University of Hawaii Cancer Center; Honolulu, HI USA
| | - Wei-Xing Zong
- Department of Molecular Genetics and Microbiology; Stony Brook University; Stony Brook, NY USA
| | - Laurence Zitvogel
- INSERM; U1015; Villejuif, France
- Gustave Roussy Cancer Campus; Villejuif, France
- Centre d’Investigation Clinique Biothérapie 507 (CICBT507); Gustave Roussy Cancer Campus; Villejuif, France
| | - Guido Kroemer
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Center de Recherche des Cordeliers; Paris, France
- INSERM; U1138; Paris, France
- Metabolomics and Cell Biology Platforms; Gustave Roussy Cancer Campus; Villejuif, France
- Université Paris Descartes/Paris V; Sorbonne Paris Cité; Paris, France
- Pôle de Biologie; Hôpital Européen Georges Pompidou; AP-HP; Paris, France
| | - Lorenzo Galluzzi
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Center de Recherche des Cordeliers; Paris, France
- INSERM; U1138; Paris, France
- Gustave Roussy Cancer Campus; Villejuif, France
- Université Paris Descartes/Paris V; Sorbonne Paris Cité; Paris, France
| |
Collapse
|
589
|
Münz C. Viral infections in mice with reconstituted human immune system components. Immunol Lett 2014; 161:118-24. [PMID: 24953718 DOI: 10.1016/j.imlet.2014.05.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Revised: 05/14/2014] [Accepted: 05/27/2014] [Indexed: 10/25/2022]
Abstract
Pathogenic viruses are often difficult to study due to their exclusive tropism for humans. The development of mice with human immune system components opens the possibility to study those human pathogens with a tropism for the human hematopoietic lineage in vivo. These include HCMV, EBV, KSHV, HIV, HTLV-1, dengue virus and JC virus. Furthermore, some human pathogens, like HSV-2, adenovirus, HCV, HBV and influenza A virus, with an additional tropism for somatic mouse tissues or for additional transplanted human tissues, mainly liver, have been explored in these models. The cellular tropism of these viruses, their associated diseases and primarily cell-mediated immune responses to these viral infections will be discussed in this review. Already some exciting information has been gained from these novel chimeric in vivo models and future avenues to gain more insights into the pathology, but also potential therapies, will be outlined. Although the respective in vivo models of human immune responses can still be significantly improved, they already provide preclinical systems for in vivo studies of important viral pathogens of humans.
Collapse
Affiliation(s)
- Christian Münz
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich, Switzerland.
| |
Collapse
|
590
|
Vudattu NK, Waldron-Lynch F, Truman LA, Deng S, Preston-Hurlburt P, Torres R, Raycroft MT, Mamula MJ, Herold KC. Humanized mice as a model for aberrant responses in human T cell immunotherapy. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2014; 193:587-96. [PMID: 24943216 PMCID: PMC4123131 DOI: 10.4049/jimmunol.1302455] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Immune-deficient mice, reconstituted with human stem cells, have been used to analyze human immune responses in vivo. Although they have been used to study immune responses to xenografts, allografts, and pathogens, there have not been models of autoimmune disease in which the mechanisms of the pathologic process can be analyzed. We have found that reconstituted "humanized" mice treated with anti-CTLA-4 Ab (ipilimumab) develop autoimmune disease characterized by hepatitis, adrenalitis, sialitis, anti-nuclear Abs, and weight loss. Induction of autoimmunity involved activation of T cells and cytokine production, and increased infiltration of APCs. When anti-CTLA-4 mAb-treated mice were cotreated with anti-CD3 mAb (teplizumab), hepatitis and anti-nuclear Abs were no longer seen and weight loss did not occur. The anti-CD3 blocked proliferation and activation of T cells, release of IFN-γ and TNF, macrophage infiltration, and release of IP-10 that was induced with anti-CTLA-4 mAb. We also found increased levels of T regulatory cells (CD25(+)CD127(-)) in the spleen and mesenteric lymph nodes in the mice treated with both Abs and greater constitutive phosphorylation of STAT5 in T regulatory cells in spleen cells compared with mice treated with anti-CTLA-4 mAb alone. We describe a model of human autoimmune disease in vivo. Humanized mice may be useful for understanding the mechanisms of biologics that are used in patients. Hepatitis, lymphadenopathy, and other inflammatory sequelae are adverse effects of ipilimumab treatment in humans, and this study may provide insights into this pathogenesis and the effects of immunologics on autoimmunity.
Collapse
MESH Headings
- Adrenal Glands/drug effects
- Adrenal Glands/immunology
- Adrenal Glands/metabolism
- Animals
- Antibodies, Monoclonal/toxicity
- Antibodies, Monoclonal, Humanized/pharmacology
- Autoimmune Diseases/chemically induced
- Autoimmune Diseases/immunology
- Autoimmune Diseases/therapy
- Cytokines/blood
- Cytokines/immunology
- Cytokines/metabolism
- Disease Models, Animal
- Flow Cytometry
- Humans
- Interleukin Receptor Common gamma Subunit/deficiency
- Interleukin Receptor Common gamma Subunit/genetics
- Ipilimumab
- Liver/drug effects
- Liver/immunology
- Liver/metabolism
- Lymphocyte Activation/drug effects
- Lymphocyte Activation/immunology
- Macrophages/drug effects
- Macrophages/immunology
- Macrophages/metabolism
- Mice
- Mice, Inbred NOD
- Mice, Knockout
- Mice, SCID
- Phosphorylation/drug effects
- Phosphorylation/immunology
- STAT5 Transcription Factor/immunology
- STAT5 Transcription Factor/metabolism
- Stem Cell Transplantation/methods
- Survival Analysis
- T-Lymphocytes/drug effects
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- T-Lymphocytes, Regulatory/drug effects
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- Transplantation, Heterologous
- Weight Loss/drug effects
- Weight Loss/immunology
Collapse
Affiliation(s)
- Nalini K Vudattu
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520
| | - Frank Waldron-Lynch
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520
| | - Lucy A Truman
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520
| | - Songyan Deng
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520
| | | | - Richard Torres
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT 06520; and
| | - Maurice T Raycroft
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520
| | - Mark J Mamula
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520
| | - Kevan C Herold
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520; Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520
| |
Collapse
|
591
|
Cosgun KN, Rahmig S, Mende N, Reinke S, Hauber I, Schäfer C, Petzold A, Weisbach H, Heidkamp G, Purbojo A, Cesnjevar R, Platz A, Bornhäuser M, Schmitz M, Dudziak D, Hauber J, Kirberg J, Waskow C. Kit regulates HSC engraftment across the human-mouse species barrier. Cell Stem Cell 2014; 15:227-38. [PMID: 25017720 DOI: 10.1016/j.stem.2014.06.001] [Citation(s) in RCA: 131] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2011] [Revised: 03/04/2014] [Accepted: 06/02/2014] [Indexed: 11/28/2022]
Abstract
In-depth analysis of the cellular and molecular mechanisms regulating human HSC function will require a surrogate host that supports robust maintenance of transplanted human HSCs in vivo, but the currently available options are problematic. Previously we showed that mutations in the Kit receptor enhance engraftment of transplanted HSCs in the mouse. To generate an improved model for human HSC transplantation and analysis, we developed immune-deficient mouse strains containing Kit mutations. We found that mutation of the Kit receptor enables robust, uniform, sustained, and serially transplantable engraftment of human HSCs in adult mice without a requirement for irradiation conditioning. Using this model, we also showed that differential KIT expression identifies two functionally distinct subpopulations of human HSCs. Thus, we have found that the capacity of this Kit mutation to open up stem cell niches across species barriers has significant potential and broad applicability in human HSC research.
Collapse
Affiliation(s)
- Kadriye Nehir Cosgun
- Regeneration in Hematopoiesis and Animal Models in Hematopoiesis, Institute for Immunology, Faculty of Medicine, TU Dresden, Fetscherstrasse 74, 01307 Dresden, Germany
| | - Susann Rahmig
- Regeneration in Hematopoiesis and Animal Models in Hematopoiesis, Institute for Immunology, Faculty of Medicine, TU Dresden, Fetscherstrasse 74, 01307 Dresden, Germany
| | - Nicole Mende
- Regeneration in Hematopoiesis and Animal Models in Hematopoiesis, Institute for Immunology, Faculty of Medicine, TU Dresden, Fetscherstrasse 74, 01307 Dresden, Germany
| | - Sören Reinke
- Regeneration in Hematopoiesis and Animal Models in Hematopoiesis, Institute for Immunology, Faculty of Medicine, TU Dresden, Fetscherstrasse 74, 01307 Dresden, Germany
| | - Ilona Hauber
- Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Martinistrasse 52, 20251 Hamburg, Germany
| | - Carola Schäfer
- Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Martinistrasse 52, 20251 Hamburg, Germany
| | - Anke Petzold
- Institute for Immunology, Faculty of Medicine, TU Dresden, Fetscherstrasse 74, 01307 Dresden, Germany
| | - Henry Weisbach
- Regeneration in Hematopoiesis and Animal Models in Hematopoiesis, Institute for Immunology, Faculty of Medicine, TU Dresden, Fetscherstrasse 74, 01307 Dresden, Germany
| | - Gordon Heidkamp
- Laboratory of Dendritic Cell Biology, Department of Dermatology, Research Module II, University Hospital of Erlangen, Friedrich-Alexander Universität Erlangen-Nürnberg, Hartmannstrasse 14, 91052 Erlangen, Germany
| | - Ariawan Purbojo
- Department of Paediatric Cardiac Surgery, University Hospital of Erlangen, Friedrich-Alexander Universität Erlangen-Nürnberg, Loschgestrasse 15, 91054 Erlangen, Germany
| | - Robert Cesnjevar
- Department of Paediatric Cardiac Surgery, University Hospital of Erlangen, Friedrich-Alexander Universität Erlangen-Nürnberg, Loschgestrasse 15, 91054 Erlangen, Germany
| | - Alexander Platz
- DKMS Lifeline Cord Blood Bank, Blasewitzer Strasse 43, 01307 Dresden, Germany
| | - Martin Bornhäuser
- Department of Hematology/Oncology, University Hospital, TU Dresden, Fetscherstr 74, 01307 Dresden, Germany
| | - Marc Schmitz
- Institute for Immunology, Faculty of Medicine, TU Dresden, Fetscherstrasse 74, 01307 Dresden, Germany
| | - Diana Dudziak
- Laboratory of Dendritic Cell Biology, Department of Dermatology, Research Module II, University Hospital of Erlangen, Friedrich-Alexander Universität Erlangen-Nürnberg, Hartmannstrasse 14, 91052 Erlangen, Germany
| | - Joachim Hauber
- Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Martinistrasse 52, 20251 Hamburg, Germany
| | - Jörg Kirberg
- Paul Ehrlich Institut, Federal Institute for Vaccines and Biomedicines, Paul-Ehrlich-Strasse 51-59, 63225 Langen, Germany
| | - Claudia Waskow
- Regeneration in Hematopoiesis and Animal Models in Hematopoiesis, Institute for Immunology, Faculty of Medicine, TU Dresden, Fetscherstrasse 74, 01307 Dresden, Germany.
| |
Collapse
|
592
|
Journal Club. Kidney Int 2014. [DOI: 10.1038/ki.2014.174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
593
|
Chen SS, Chiorazzi N. Murine genetically engineered and human xenograft models of chronic lymphocytic leukemia. Semin Hematol 2014; 51:188-205. [PMID: 25048783 DOI: 10.1053/j.seminhematol.2014.05.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Chronic lymphocytic leukemia (CLL) is a genetically complex disease, with multiple factors having an impact on onset, progression, and response to therapy. Genetic differences/abnormalities have been found in hematopoietic stem cells from patients, as well as in B lymphocytes of individuals with monoclonal B-cell lymphocytosis who may develop the disease. Furthermore, after the onset of CLL, additional genetic alterations occur over time, often causing disease worsening and altering patient outcomes. Therefore, being able to genetically engineer mouse models that mimic CLL or at least certain aspects of the disease will help us understand disease mechanisms and improve treatments. This notwithstanding, because neither the genetic aberrations responsible for leukemogenesis and progression nor the promoting factors that support these are likely identical in character or influences for all patients, genetically engineered mouse models will only completely mimic CLL when all of these factors are precisely defined. In addition, multiple genetically engineered models may be required because of the heterogeneity in susceptibility genes among patients that can have an effect on genetic and environmental characteristics influencing disease development and outcome. For these reasons, we review the major murine genetically engineered and human xenograft models in use at the present time, aiming to report the advantages and disadvantages of each.
Collapse
Affiliation(s)
- Shih-Shih Chen
- The Feinstein Institute for Medical Research, North Shore-LIJ Health System, Manhasset, New York.
| | - Nicholas Chiorazzi
- The Feinstein Institute for Medical Research, North Shore-LIJ Health System, Manhasset, New York; Departments of Medicine and Molecular Medicine, Hofstra North Shore-LIJ School of Medicine, Manhasset, New York.
| |
Collapse
|
594
|
Affiliation(s)
- Hergen Spits
- Department of Cell Biology and Histology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
595
|
Leavy O. Mice with a human innate compartment. Nat Rev Immunol 2014. [DOI: 10.1038/nri3654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|