201
|
Herati RS, Wherry EJ. What Is the Predictive Value of Animal Models for Vaccine Efficacy in Humans? Consideration of Strategies to Improve the Value of Animal Models. Cold Spring Harb Perspect Biol 2018; 10:cshperspect.a031583. [PMID: 28348037 DOI: 10.1101/cshperspect.a031583] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Animal models are an essential feature of the vaccine design toolkit. Although animal models have been invaluable in delineating the mechanisms of immune function, their precision in predicting how well specific vaccines work in humans is often suboptimal. There are, of course, many obvious species differences that may limit animal models from predicting all details of how a vaccine works in humans. However, careful consideration of which animal models may have limitations should also allow more accurate interpretations of animal model data and more accurate predictions of what is to be expected in clinical trials. In this article, we examine some of the considerations that might be relevant to cross-species extrapolation of vaccine-related immune responses for the prediction of how vaccines will perform in humans.
Collapse
Affiliation(s)
- Ramin Sedaghat Herati
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104.,Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104
| | - E John Wherry
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104.,Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104
| |
Collapse
|
202
|
Clatworthy AE, Romano KP, Hung DT. Whole-organism phenotypic screening for anti-infectives promoting host health. Nat Chem Biol 2018; 14:331-341. [PMID: 29556098 PMCID: PMC9843822 DOI: 10.1038/s41589-018-0018-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Accepted: 11/20/2017] [Indexed: 01/19/2023]
Abstract
To date, antibiotics have been identified on the basis of their ability to kill bacteria or inhibit their growth rather than directly for their capacity to improve clinical outcomes of infected patients. Although historically successful, this approach has led to the development of an antibiotic armamentarium that suffers from a number of shortcomings, including the inevitable emergence of resistance and, in certain infections, suboptimal efficacy leading to long treatment durations, infection recurrence, or high mortality and morbidity rates despite apparent bacterial sterilization. Conventional antibiotics fail to address the complexities of in vivo bacterial physiology and virulence, as well as the role of the host underlying the complex, dynamic interactions that cause disease. New interventions are needed, aimed at host outcome rather than microbiological cure. Here we review the role of screening models for cellular and whole-organism infection, including worms, flies, zebrafish, and mice, to identify novel therapeutic strategies and discuss their future implications.
Collapse
Affiliation(s)
- Anne E. Clatworthy
- Broad Institute of MIT and Harvard, Cambridge, MA, USA,Department of Molecular Biology and Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, MA, USA,Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Keith P. Romano
- Department of Molecular Biology and Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, MA, USA,Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| | - Deborah T. Hung
- Broad Institute of MIT and Harvard, Cambridge, MA, USA,Department of Molecular Biology and Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, MA, USA,Department of Genetics, Harvard Medical School, Boston, MA, USA,Correspondence and requests for materials should be addressed to D.T.H.
| |
Collapse
|
203
|
Singer BH, Dickson RP, Denstaedt SJ, Newstead MW, Kim K, Falkowski NR, Erb-Downward JR, Schmidt TM, Huffnagle GB, Standiford TJ. Bacterial Dissemination to the Brain in Sepsis. Am J Respir Crit Care Med 2018; 197:747-756. [PMID: 29232157 PMCID: PMC5855074 DOI: 10.1164/rccm.201708-1559oc] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Accepted: 12/12/2017] [Indexed: 12/11/2022] Open
Abstract
RATIONALE Sepsis causes brain dysfunction and neuroinflammation. It is unknown whether neuroinflammation in sepsis is initiated by dissemination of bacteria to the brain and sustained by persistent infection, or whether neuroinflammation is a sterile process resulting solely from circulating inflammatory mediators. OBJECTIVES To determine if gut bacteria translocate to the brain during sepsis, and are associated with neuroinflammation. METHODS Murine sepsis was induced using cecal ligation and puncture, and sepsis survivor mice were compared with sham and unoperated control animals. Brain tissue of patients who died of sepsis was compared with patients who died of noninfectious causes. Bacterial taxa were characterized by 16S ribosomal RNA gene sequencing in both murine and human brain specimens; compared among sepsis and nonsepsis groups; and correlated with levels of S100A8, a marker of neuroinflammation using permutational multivariate ANOVA. MEASUREMENTS AND MAIN RESULTS Viable gut-associated bacteria were enriched in the brains of mice 5 days after surviving abdominal sepsis (P < 0.01), and undetectable by 14 days. The community structure of brain-associated bacteria correlated with severity of neuroinflammation (P < 0.001). Furthermore, bacterial taxa detected in brains of humans who die of sepsis were distinct from those who died of noninfectious causes (P < 0.001) and correlated with S100A8/A9 expression (P < 0.05). CONCLUSIONS Although bacterial translocation is associated with acute neuroinflammation in murine sepsis, bacterial translocation did not result in chronic cerebral infection. Postmortem analysis of patients who die of sepsis suggests a role for bacteria in acute brain dysfunction in sepsis. Further work is needed to determine if modifying gut-associated bacterial communities modulates brain dysfunction after sepsis.
Collapse
Affiliation(s)
- Benjamin H. Singer
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, and
- Michigan Center for Integrative Research in Critical Care, Ann Arbor, Michigan
| | - Robert P. Dickson
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, and
- Michigan Center for Integrative Research in Critical Care, Ann Arbor, Michigan
| | - Scott J. Denstaedt
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, and
| | - Michael W. Newstead
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, and
| | - Kwi Kim
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan; and
| | - Nicole R. Falkowski
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, and
| | - John R. Erb-Downward
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, and
| | - Thomas M. Schmidt
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan; and
| | - Gary B. Huffnagle
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, and
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan; and
| | | |
Collapse
|
204
|
Serve R, Sturm R, Schimunek L, Störmann P, Heftrig D, Teuben MPJ, Oppermann E, Horst K, Pfeifer R, Simon TP, Kalbas Y, Pape HC, Hildebrand F, Marzi I, Relja B. Comparative Analysis of the Regulatory T Cells Dynamics in Peripheral Blood in Human and Porcine Polytrauma. Front Immunol 2018; 9:435. [PMID: 29593715 PMCID: PMC5859958 DOI: 10.3389/fimmu.2018.00435] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Accepted: 02/19/2018] [Indexed: 12/18/2022] Open
Abstract
Background Severely injured patients experience substantial immunological stress in the aftermath of traumatic insult, which often results in systemic immune dysregulation. Regulatory T cells (Treg) play a key role in the suppression of the immune response and in the maintenance of immunological homeostasis. Little is known about their presence and dynamics in blood after trauma, and nothing is known about Treg in the porcine polytrauma model. Here, we assessed different subsets of Treg in trauma patients (TP) and compared those to either healthy volunteers (HV) or data from porcine polytrauma. Methods Peripheral blood was withdrawn from 20 TP with injury severity score (ISS) ≥16 at the admittance to the emergency department (ED), and subsequently on day 1 and at day 3. Ten HV were included as controls (ctrl). The porcine polytrauma model consisted of a femur fracture, liver laceration, lung contusion, and hemorrhagic shock resulting in an ISS of 27. After polytrauma, the animals underwent resuscitation and surgical fracture fixation. Blood samples were withdrawn before and immediately after trauma, 24 and 72 h later. Different subsets of Treg, CD4+CD25+, CD4+CD25+FoxP3+, CD4+CD25+CD127-, and CD4+CD25+CD127-FoxP3+ were characterized by flow cytometry. Results Absolute cell counts of leukocytes were significantly increasing after trauma, and again decreasing in the follow-up in human and porcine samples. The proportion of human Treg in the peripheral blood of TP admitted to the ED was lower when compared to HV. Their numbers did not recover until 72 h after trauma. Comparable data were found for all subsets. The situation in the porcine trauma model was comparable with the clinical data. In porcine peripheral blood before trauma, we could identify Treg with the typical immunophenotype (CD4+CD25+CD127-), which were virtually absent immediately after trauma. Similar to the human situation, most of these cells expressed FoxP3, as assessed by intracellular FACS stain. Conclusion Despite minor percental differences in the recovery of Treg populations after trauma, our findings show a comparable decrease of Treg early after polytrauma, and strengthen the immunological significance of the porcine polytrauma model. Furthermore, the Treg subpopulation CD4+CD25+CD127- was characterized in porcine samples.
Collapse
Affiliation(s)
- Rafael Serve
- Department of Trauma, Hand and Reconstructive Surgery, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | - Ramona Sturm
- Department of Trauma, Hand and Reconstructive Surgery, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | - Lukas Schimunek
- Department of Trauma, Hand and Reconstructive Surgery, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | - Philipp Störmann
- Department of Trauma, Hand and Reconstructive Surgery, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | - David Heftrig
- Department of Trauma, Hand and Reconstructive Surgery, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | - Michel P. J. Teuben
- Department of Orthopaedic Trauma Surgery, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Elsie Oppermann
- Department of Abdominal and Visceral Surgery, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | - Klemens Horst
- Department of Orthopaedic Trauma, RWTH Aachen University, Aachen, Germany
| | - Roman Pfeifer
- Department of Orthopaedic Trauma Surgery, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Tim P. Simon
- Department of Intensive Care and Intermediate Care, RWTH Aachen University, Aachen, Germany
| | - Yannik Kalbas
- Department of Orthopaedic Trauma Surgery, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Hans-Christoph Pape
- Department of Orthopaedic Trauma Surgery, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Frank Hildebrand
- Department of Orthopaedic Trauma, RWTH Aachen University, Aachen, Germany
| | - Ingo Marzi
- Department of Trauma, Hand and Reconstructive Surgery, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | - Borna Relja
- Department of Trauma, Hand and Reconstructive Surgery, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| |
Collapse
|
205
|
Peter A, Kovarova M, Staiger H, Machann J, Schick F, Königsrainer A, Königsrainer I, Schleicher E, Fritsche A, Häring HU, Stefan N. The hepatokines fetuin-A and fetuin-B are upregulated in the state of hepatic steatosis and may differently impact on glucose homeostasis in humans. Am J Physiol Endocrinol Metab 2018; 314:E266-E273. [PMID: 29138227 DOI: 10.1152/ajpendo.00262.2017] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The liver is a central regulator of whole body glucose, and lipid homeostasis and hepatokines, like fetuin-A, have been identified as markers and mediators of fatty liver-induced cardiometabolic risk. The closely related protein fetuin-B was shown to be upregulated in the fatty liver and to impact on glucose homeostasis in mice. In the present study we aimed to test the relevance of these findings in humans. In 55 subjects, hepatic mRNA expression of both hepatokines, fetuin-A and fetuin-B, associated positively with liver triglyceride content, whereas only fetuin-A expression associated with the homeostatic model assessment of insulin resistance. In 220 subjects who underwent precise metabolic phenotyping, circulating fetuin-A, but not fetuin-B, associated positively with liver fat content, and negatively with insulin sensitivity, measured during the oral glucose tolerance test (OGTT) and during the euglycemic, hyperinsulinemic clamp. Both circulating fetuin-A and fetuin-B correlated positively with the glucose area under the curve during the OGTT, but after additional adjustment for insulin sensitivity this relationship remained significant only for fetuin-B. In conclusion, despite the fact that the two hepatokines, fetuin-A and fetuin-B, are upregulated in the state of hepatic steatosis in humans, it appears that they differently impact on glucose homeostasis. Our data are in agreement with observations that fetuin-A can alter insulin signaling and that fetuin-B may regulate glucose homeostasis via so far unknown effects, possibly on glucose effectiveness.
Collapse
Affiliation(s)
- Andreas Peter
- Department of Internal Medicine IV, Division of Endocrinology, Diabetology, Vascular Medicine, Nephrology and Clinical Chemistry, University of Tübingen , Tübingen , Germany
- Institute for Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Center Munich at the University of Tübingen , Tübingen , Germany
- German Center for Diabetes Research (DZD), München- Neuherberg , Germany
| | - Marketa Kovarova
- Department of Internal Medicine IV, Division of Endocrinology, Diabetology, Vascular Medicine, Nephrology and Clinical Chemistry, University of Tübingen , Tübingen , Germany
| | - Harald Staiger
- Institute for Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Center Munich at the University of Tübingen , Tübingen , Germany
- German Center for Diabetes Research (DZD), München- Neuherberg , Germany
- Institute for Pharmaceutical Sciences, Department of Pharmacy and Biochemistry, University of Tübingen , Tübingen , Germany
| | - Jürgen Machann
- Institute for Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Center Munich at the University of Tübingen , Tübingen , Germany
- German Center for Diabetes Research (DZD), München- Neuherberg , Germany
| | - Fritz Schick
- Institute for Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Center Munich at the University of Tübingen , Tübingen , Germany
- German Center for Diabetes Research (DZD), München- Neuherberg , Germany
- Section on Experimental Radiology, University of Tübingen , Tübingen , Germany
| | - Alfred Königsrainer
- Department of General, Visceral and Transplant Surgery, University of Tübingen , Tübingen , Germany
| | - Ingmar Königsrainer
- Department of General, Visceral and Transplant Surgery, University of Tübingen , Tübingen , Germany
| | - Erwin Schleicher
- Department of Internal Medicine IV, Division of Endocrinology, Diabetology, Vascular Medicine, Nephrology and Clinical Chemistry, University of Tübingen , Tübingen , Germany
- Institute for Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Center Munich at the University of Tübingen , Tübingen , Germany
- German Center for Diabetes Research (DZD), München- Neuherberg , Germany
| | - Andreas Fritsche
- Department of Internal Medicine IV, Division of Endocrinology, Diabetology, Vascular Medicine, Nephrology and Clinical Chemistry, University of Tübingen , Tübingen , Germany
- Institute for Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Center Munich at the University of Tübingen , Tübingen , Germany
- German Center for Diabetes Research (DZD), München- Neuherberg , Germany
| | - Hans-Ulrich Häring
- Department of Internal Medicine IV, Division of Endocrinology, Diabetology, Vascular Medicine, Nephrology and Clinical Chemistry, University of Tübingen , Tübingen , Germany
- Institute for Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Center Munich at the University of Tübingen , Tübingen , Germany
- German Center for Diabetes Research (DZD), München- Neuherberg , Germany
| | - Norbert Stefan
- Department of Internal Medicine IV, Division of Endocrinology, Diabetology, Vascular Medicine, Nephrology and Clinical Chemistry, University of Tübingen , Tübingen , Germany
- Institute for Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Center Munich at the University of Tübingen , Tübingen , Germany
- German Center for Diabetes Research (DZD), München- Neuherberg , Germany
| |
Collapse
|
206
|
Moore BB. Groundhog Day for Rodent Models of Acute Lung Injury: Clear Relevance or Renewed Debate? Am J Respir Cell Mol Biol 2018; 57:141-142. [PMID: 28762770 DOI: 10.1165/rcmb.2017-0119ed] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Affiliation(s)
- Bethany B Moore
- 1 Department of Internal Medicine University of Michigan Ann Arbor, Michigan
| |
Collapse
|
207
|
Abstract
The molecular basis responsible for tolerance following inflammatory response to lipopolysaccharide (LPS) is not well understood. We hypothesized that inflammation/tolerance in monocytes/ macrophages is dependent on the proteases of proteasome. To test our hypothesis, first, we examined the expression of different proteasome subunits in different human and mouse monocytes/macrophages. Secondly, we investigated the effect of proteasome subunits/ proteases on LPS-induced expression of tumor necrosis factor-α (TNF-α) and nitric oxide (NO) during inflammation and tolerance using mouse RAW 264.7 macrophages, THP1 cells, and cluster of differentiation 14 positive (CD14) human monocytes. We found that RAW 264.7 cells (XYZ), mouse peritoneal resident, thioglycollate-elicited macrophages, primed RAW 264.7 (XYZ, LMP), and human monocytes (LMP) expressed different types of proteasome subunits/activities. Cells containing predominantly either LMP subunits (such as THP-1 and human monocytes), or only X, Y, Z subunits (RAW 264.7 cells not primed) could only induce TNF-α, but not NO, while cells containing all five to six subunits (XYZ, LMP) of the proteasome could induce both mediators in response to LPS. Distinct states of inflammation/tolerance in LPS treated cells, strongly correlated with an upregulation or downregulation of proteasome's subunits (proteases), respectively. Moreover, interferon-γ treatment of tolerant cells caused robust induction of proteasome's subunit expression in mouse macrophages and human monocytes, and cells regained their ability to respond to LPS. These studies are vital for understanding function of proteasome's subunits during inflammation/tolerance in mouse and human cells, and for design of therapeutic strategies for all diseases based on inflammation.
Collapse
|
208
|
Löwa A, Jevtić M, Gorreja F, Hedtrich S. Alternatives to animal testing in basic and preclinical research of atopic dermatitis. Exp Dermatol 2018; 27:476-483. [PMID: 29356091 DOI: 10.1111/exd.13498] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/12/2018] [Indexed: 12/29/2022]
Abstract
Atopic dermatitis (AD) is a chronic inflammatory skin disease of increasing prevalence, especially in industrialized countries. Roughly 25% of the children and 1%-3% of adults are affected. Although significant progress has been made in the understanding of the pathogenesis of AD, many aspects remain poorly understood. Moreover, there is a pressing need for improved therapeutic options. Studies to elucidate the pathophysiological pathways of AD and to identify novel therapeutic targets over the last few decades have been conducted almost exclusively in animal models. However, in vitro approaches such as 3D skin disease models have recently emerged due to an increasing awareness of distinct interspecies-related differences that hamper the effective translation of results from animal models to humans. In addition, there is growing political and social pressure to develop alternatives to animal models according to the 3Rs principle (reduction, refinement and replacement of animal models).
Collapse
Affiliation(s)
- Anna Löwa
- Institute for Pharmacy, Pharmacology & Toxicology, Freie Universität Berlin, Berlin, Germany
| | - Marijana Jevtić
- Institute for Pharmacy, Pharmacology & Toxicology, Freie Universität Berlin, Berlin, Germany
| | - Frida Gorreja
- Örebro University, School of Health and Medical Sciences, Örebro University, Orebro, Sweden
| | - Sarah Hedtrich
- Institute for Pharmacy, Pharmacology & Toxicology, Freie Universität Berlin, Berlin, Germany
| |
Collapse
|
209
|
Lewis AJ, Rosengart MR. Bench-to-Bedside: A Translational Perspective on Murine Models of Sepsis. Surg Infect (Larchmt) 2018; 19:137-141. [PMID: 29394153 PMCID: PMC5815447 DOI: 10.1089/sur.2017.308] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Considerable research effort has focused on the development of novel therapies for the treatment of sepsis, yet after decades of clinical trials, few significant advances have been achieved. This limitation persists despite a wealth of data yielded by basic science that has expanded our knowledge of the biology of this disease exponentially. METHOD Review of the English-language literature. RESULTS Translational researchers may address the resultant gap between the basic science laboratory and clinical research worlds. Herein, we review potential causes for the challenges of translating basic laboratory discovery into clinical benefit. CONCLUSION We propose conceptual platforms to further the development of translational sepsis research efforts.
Collapse
Affiliation(s)
- Anthony J. Lewis
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Matthew R. Rosengart
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
210
|
Smith TJ. New advances in understanding thyroid-associated ophthalmopathy and the potential role for insulin-like growth factor-I receptor. F1000Res 2018; 7:134. [PMID: 29744034 PMCID: PMC5795270 DOI: 10.12688/f1000research.12787.1] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/24/2018] [Indexed: 01/15/2023] Open
Abstract
Thyroid-associated ophthalmopathy (TAO), a localized periocular manifestation of the autoimmune syndrome known as Graves’ disease, remains incompletely understood. Discussions of its pathogenesis are generally focused on the thyrotropin receptor, the proposed role for which is supported by substantial evidence. Considerations of any involvement of the insulin-like growth factor-I receptor (IGF-IR) in the disease are frequently contentious. In this brief, topically focused review, I have attempted to provide a balanced perspective based entirely on experimental results that either favor or refute involvement of IGF-IR in TAO. Discussion in this matter seems particularly timely since the currently available treatments of this disfiguring and potentially sight-threatening disease remain inadequate. Importantly, no medical therapy has thus far received approval from the US Food and Drug Administration. Results from a very recently published clinical trial assessing the safety and efficacy of teprotumumab, an inhibitory human anti–IGF-IR monoclonal antibody, in active, moderate to severe TAO are extremely encouraging. That double-masked, placebo-controlled study involved 88 patients and revealed unprecedented clinical responses in the improvement of proptosis and clinical activity as well as a favorable safety profile. Should those results prove reproducible in an ongoing phase III trial, therapeutic inhibition of IGF-IR could become the basis for paradigm-shifting treatment of this vexing disease.
Collapse
Affiliation(s)
- Terry J Smith
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center and Division of Metabolism, Endocrinology and Diabetes, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48105, USA
| |
Collapse
|
211
|
Effect of hyperbaric oxygen therapy (HBO) on implant-associated osteitis in a femur fracture model in mice. PLoS One 2018; 13:e0191594. [PMID: 29377928 PMCID: PMC5788341 DOI: 10.1371/journal.pone.0191594] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 01/08/2018] [Indexed: 11/19/2022] Open
Abstract
Hyperbaric oxygen therapy (HBO) is applied very successfully in treatment of various diseases such as chronic wounds. It has been already suggested as adjunctive treatment option for osteitis by immune- and fracture modulating effects. This study evaluates the importance of HBO in an early implant-associated localized osteitis caused by Staphylococcus aureus (SA) compared to the standard therapy. In a standardized murine model the left femur of 120 BALB/c mice were osteotomized and fixed by a titanium locking plate. Osteitis has been induced with a defined amount of SA into the fracture gap. Debridément and lavages were progressed on day 7, 14, 28 and 56 to determine the local bacterial growth and the immune reaction. Hyperbaric oxygen (2 ATA, 90%) was applied for 90 minutes on day 7 to 21 for those mice allocated to HBO therapy. To evaluate the effect of HBO therapy the following groups were analyzed: Two sham-groups (12 mice / group) with and without HBO therapy, two osteotomy groups (24 mice / group) with plate osteosynthesis of the femur with and without HBO therapy, and two osteotomy SA infection groups (24 mice / group) with and without HBO therapy. Fracture healing was also quantified on day 7, 14, 28 and 56 by a.p. x-ray and bone healing markers from blood samples. Progression of infection was assessed by estimation of colony-forming units (CFU) and immune response was analyzed by determination of polymorphonuclear neutrophils (PMN), Interleukin (IL) - 6, and the circulating free DNA (cfDNA) in lavage samples. Osteitis induced significantly higher IL-6, cfDNA- and PMN-levels in the lavage samples (on day 7 and 14, each p < 0.05). HBO-therapy did not have a significant influence on the CFU and immune response compared to the standard therapy (each p > 0.05). At the same time HBO-therapy was associated with a delayed bone healing assessed by x-ray radiography and a higher rate of non-union until day 28. In conclusion, osteitis led to significantly higher bacterial count and infection parameters. HBO-therapy neither had a beneficial influence on local infection nor on immune response or fracture healing compared to the standard therapy in an osteitis mouse model.
Collapse
|
212
|
Zscheppang K, Berg J, Hedtrich S, Verheyen L, Wagner DE, Suttorp N, Hippenstiel S, Hocke AC. Human Pulmonary 3D Models For Translational Research. Biotechnol J 2018; 13:1700341. [PMID: 28865134 PMCID: PMC7161817 DOI: 10.1002/biot.201700341] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 08/23/2017] [Indexed: 12/13/2022]
Abstract
Lung diseases belong to the major causes of death worldwide. Recent innovative methodological developments now allow more and more for the use of primary human tissue and cells to model such diseases. In this regard, the review covers bronchial air-liquid interface cultures, precision cut lung slices as well as ex vivo cultures of explanted peripheral lung tissue and de-/re-cellularization models. Diseases such as asthma or infections are discussed and an outlook on further areas for development is given. Overall, the progress in ex vivo modeling by using primary human material could make translational research activities more efficient by simultaneously fostering the mechanistic understanding of human lung diseases while reducing animal usage in biomedical research.
Collapse
Affiliation(s)
- Katja Zscheppang
- Dept. of Internal Medicine/Infectious and Respiratory DiseasesCharité − Universitätsmedizin BerlinCharitèplatz 1Berlin 10117Germany
| | - Johanna Berg
- Department of BiotechnologyTechnical University of BerlinGustav‐Meyer‐Allee 25Berlin 13335Germany
| | - Sarah Hedtrich
- Institute for PharmacyPharmacology and ToxicologyFreie Universität BerlinBerlinGermany
| | - Leonie Verheyen
- Institute for PharmacyPharmacology and ToxicologyFreie Universität BerlinBerlinGermany
| | - Darcy E. Wagner
- Helmholtz Zentrum Munich, Lung Repair and Regeneration Unit, Comprehensive Pneumology CenterMember of the German Center for Lung ResearchMunichGermany
| | - Norbert Suttorp
- Dept. of Internal Medicine/Infectious and Respiratory DiseasesCharité − Universitätsmedizin BerlinCharitèplatz 1Berlin 10117Germany
| | - Stefan Hippenstiel
- Dept. of Internal Medicine/Infectious and Respiratory DiseasesCharité − Universitätsmedizin BerlinCharitèplatz 1Berlin 10117Germany
| | - Andreas C. Hocke
- Dept. of Internal Medicine/Infectious and Respiratory DiseasesCharité − Universitätsmedizin BerlinCharitèplatz 1Berlin 10117Germany
| |
Collapse
|
213
|
Mira JC, Nacionales DC, Loftus TJ, Ungaro R, Mathias B, Mohr AM, Moldawer LL, Efron PA. Mouse Injury Model of Polytrauma and Shock. Methods Mol Biol 2018; 1717:1-15. [PMID: 29468579 PMCID: PMC6296232 DOI: 10.1007/978-1-4939-7526-6_1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Severe injury and shock remain major sources of morbidity and mortality worldwide. Immunologic dysregulation following trauma contributes to these poor outcomes. Few, if any, therapeutic interventions have benefited these patients, and this is due to our limited understanding of the host response to injury and shock. The Food and Drug Administration requires preclinical animal studies prior to any interventional trials in humans; thus, animal models of injury and shock will remain the mainstay for trauma research. However, adequate animal models that reflect the severe response to trauma in both the acute and subacute phases have been limited. Here we describe a novel murine model of polytrauma and shock that combines hemorrhagic shock, cecectomy, long bone fracture, and soft-tissue damage. This model produces an equivalent Injury Severity Score associated with adverse outcomes in humans, and may better recapitulate the human leukocyte, cytokine, transcriptomic, and overall inflammatory response following injury and hemorrhagic shock.
Collapse
Affiliation(s)
- Juan C Mira
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, USA
| | - Dina C Nacionales
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, USA
| | - Tyler J Loftus
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, USA
| | - Ricardo Ungaro
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, USA
| | - Brittany Mathias
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, USA
| | - Alicia M Mohr
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, USA
| | - Lyle L Moldawer
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, USA
| | - Philip A Efron
- Department of Surgery, Shands Hospital, University of Florida College of Medicine, Gainesville, FL, USA.
| |
Collapse
|
214
|
Gregory DJ, Kramnik I, Kobzik L. Protection of macrophages from intracellular pathogens by miR-182-5p mimic-a gene expression meta-analysis approach. FEBS J 2017; 285:244-260. [PMID: 29197182 DOI: 10.1111/febs.14348] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Revised: 09/29/2017] [Accepted: 11/28/2017] [Indexed: 12/25/2022]
Abstract
The goals of this study were to (a) define which host genes are of particular importance during the interactions between macrophages and intracellular pathogens, and (b) use this knowledge to gain fresh, experimental understanding of how macrophage activities may be manipulated during host defense. We designed an in silico method for meta-analysis of microarray gene expression data, and used this to combine data from 16 different studies of cells in the monocyte-macrophage lineage infected with seven different pathogens. Three thousand four hundred ninety-eight genes were identified, which we call the macrophage intracellular pathogen response (macIPR) gene set. As expected, the macIPR gene set showed a strong bias toward genes previously associated with the immune response. Predicted target sites for miR-182-5p (miR-182) were strongly over-represented among macIPR genes, indicating an unexpected role for miR-182-regulatable genes during intracellular pathogenesis. We therefore transfected primary human alveolar macrophage-like monocyte-derived macrophages from multiple different donors with synthetic miR-182, and found that miR-182 overexpression (a) increases proinflammatory gene induction during infection with Francisella tularensis live vaccine strain (LVS), (b) primes macrophages for increased autophagy, and (c) enhances macrophage control of both gram negative F. tularensisLVS and gram positive Bacillus anthracisANR-1 spores. These data therefore suggest a new application for miR-182 in promoting resistance to intracellular pathogens.
Collapse
Affiliation(s)
- David J Gregory
- Molecular and Physiological Sciences Program, Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Igor Kramnik
- Pulmonary Center, Department of Medicine, National Emerging Infectious Diseases Laboratories, Boston University School of Medicine, MA, USA
| | - Lester Kobzik
- Molecular and Physiological Sciences Program, Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| |
Collapse
|
215
|
Benson RA, McInnes IB, Garside P, Brewer JM. Model answers: Rational application of murine models in arthritis research. Eur J Immunol 2017; 48:32-38. [PMID: 29193037 PMCID: PMC5814907 DOI: 10.1002/eji.201746938] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 11/02/2017] [Accepted: 11/22/2017] [Indexed: 12/29/2022]
Abstract
Advances in targeted immune therapeutics have profoundly improved clinical outcomes for patients with inflammatory arthropathies particularly rheumatoid arthritis. The landscape of disease that is observed and the treatment outcomes desired for the future have also progressed. As such there is an increasing move away from traditional models of end‐stage, chronic disease with recognition of the need to consider the earliest phases of pathogenesis as a target for treatment leading to resolution and/or cure. In order to continue the discovery process and enhance our understanding of disease and treatment, we therefore need to continuously revisit the animal models we employ and assess their relevance and utility in the light of contemporary therapeutic goals. In this review, we highlight the areas where we consider new developments in animal models and their application are most required. Thus, we have contextualised the relevant mouse models and their use within the current concepts of human inflammatory arthritis pathogenesis and highlight areas of need.
Collapse
Affiliation(s)
- Robert A Benson
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary & Life Sciences, Sir Graeme Davies Building, University of Glasgow, Glasgow, UK
| | - Iain B McInnes
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary & Life Sciences, Sir Graeme Davies Building, University of Glasgow, Glasgow, UK
| | - Paul Garside
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary & Life Sciences, Sir Graeme Davies Building, University of Glasgow, Glasgow, UK
| | - James M Brewer
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary & Life Sciences, Sir Graeme Davies Building, University of Glasgow, Glasgow, UK
| |
Collapse
|
216
|
Danahy DB, Strother RK, Badovinac VP, Griffith TS. Clinical and Experimental Sepsis Impairs CD8 T-Cell-Mediated Immunity. Crit Rev Immunol 2017; 36:57-74. [PMID: 27480902 DOI: 10.1615/critrevimmunol.2016017098] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Septic patients experience chronic immunosuppression resulting in enhanced susceptibility to infections normally controlled by T cells. Clinical research on septic patients has shown increased apoptosis and reduced total numbers of CD4 and CD8 T cells, suggesting contributing mechanism driving immunosuppression. Experimental models of sepsis, including cecal ligation and puncture, reverse translated this clinical observation to facilitate hypothesis-driven research and allow the use of an array of experimental tools to probe the impact of sepsis on T-cell immunity. In addition to numerical loss, sepsis functionally impairs the antigen-driven proliferative capacity and effector functions of CD4 and CD8 T cells. Sepsis-induced impairments in both the quantity and quality of T cells results in reduced protective capacity and increased susceptibility of mice to new or previously encountered infections. Therefore, the combined efforts of clinical and experimental sepsis research have begun to elucidate the impact of sepsis on T-cell-mediated immunity and potential T-cell-intrinsic and -extrinsic mechanisms driving chronic immunosuppression. Future work will explore the impact of sepsis on the recently appreciated tissue-resident memory (TRM) T cells, which provide robust protection against localized infections, and dendritic cells, which are needed to activate T cells and promote effective T-cell responses.
Collapse
Affiliation(s)
- Derek B Danahy
- Department of Pathology, University of Iowa, Iowa City, IA; Interdisciplinary Program in Immunology, University of Iowa, Iowa City, IA
| | | | - Vladimir P Badovinac
- Department of Pathology, Interdisciplinary Program in Immunology, University of Iowa, Iowa City, Iowa
| | - Thomas S Griffith
- Department of Urology, University of Minnesota, Minneapolis, MN; Microbiology, Immunology and Cancer Biology Graduate Program, University of Minnesota, Minneapolis, MN; Center for Immunology, University of Minnesota, Minneapolis, MN; Minneapolis VA Health Care System, Minneapolis, Minnesota
| |
Collapse
|
217
|
Modeling Acute Traumatic Hemorrhagic Shock Injury: Challenges and Guidelines for Preclinical Studies. Shock 2017; 48:610-623. [DOI: 10.1097/shk.0000000000000901] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
|
218
|
Joachim R, Suber F, Kobzik L. Characterising Pre-pubertal Resistance to Death from Endotoxemia. Sci Rep 2017; 7:16541. [PMID: 29185479 PMCID: PMC5707402 DOI: 10.1038/s41598-017-16743-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 11/15/2017] [Indexed: 02/06/2023] Open
Abstract
Sepsis is a common and deadly syndrome in which a dysregulated host response to infection causes organ failure and death. The current lack of treatment options suggests that a new approach to studying sepsis is needed. Pre-pubertal children show a relative resistance to death from severe infections and sepsis. To explore this phenomenon experimentally, we used an endotoxemia model of sepsis in mice. Following intra-peritoneal injection of endotoxin, pre-pubertal mice showed greater survival than post-pubertal mice (76.3% vs. 28.6%), despite exhibiting a similar degree of inflammation after two hours. Age-associated differences in the inflammatory response only became evident at twenty hours, when post-pubertal mice showed prolonged elevation of serum cytokines and differential recruitment of peritoneal immune cells. Mechanistically, prevention of puberty by hormonal blockade or acceleration of puberty by oestrogen treatment led to increased or decreased survival from endotoxemia, respectively. Additionally, the adoptive transfer of pre-pubertal peritoneal cells improved the survival of post-pubertal recipient mice, while post-pubertal peritoneal cells or vehicle did not. These data establish a model for studying childhood resistance to mortality from endotoxemia, demonstrate that oestrogen is responsible for an increased susceptibility to mortality after puberty, and identify peritoneal cells as mediators of pre-pubertal resistance.
Collapse
Affiliation(s)
- Rose Joachim
- Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Freeman Suber
- Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Lester Kobzik
- Harvard T. H. Chan School of Public Health, Boston, MA, USA.
| |
Collapse
|
219
|
Abstract
Safe and efficacious vaccines are arguably the most successful medical interventions of all time. Yet the ongoing discovery of new pathogens, along with emergence of antibiotic-resistant pathogens and a burgeoning population at risk of such infections, imposes unprecedented public health challenges. To meet these challenges, innovative strategies to discover and develop new or improved anti-infective vaccines are necessary. These approaches must intersect the most meaningful insights into protective immunity and advanced technologies with capabilities to deliver immunogens for optimal immune protection. This goal is considered through several recent advances in host-pathogen relationships, conceptual strides in vaccinology, and emerging technologies. Given a clear and growing risk of pandemic disease should the threat of infection go unmet, developing vaccines that optimize protective immunity against high-priority and antibiotic-resistant pathogens represents an urgent and unifying imperative.
Collapse
Affiliation(s)
- Michael R Yeaman
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California 90024.,Division of Molecular Medicine, Department of Medicine, Harbor-UCLA Medical Center, Torrance, California 90509; .,Division of Infectious Diseases, Department of Medicine, Harbor-UCLA Medical Center, Torrance, California 90509.,Los Angeles Biomedical Research Institute, Torrance, California 90502
| | | |
Collapse
|
220
|
Bolker JA. Animal Models in Translational Research: Rosetta Stone or Stumbling Block? Bioessays 2017; 39. [PMID: 29052843 DOI: 10.1002/bies.201700089] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 09/17/2017] [Indexed: 01/12/2023]
Abstract
Leading animal models are powerful tools for translational research, but they also present obstacles. Poorly conducted preclinical research in animals is a common cause of translational failure, but even when such research is well-designed and carefully executed, challenges remain. In particular, dominant models may bias research directions, elide essential aspects of human disease, omit important context, or subtly shift research targets. Recognizing these stumbling blocks can help us find ways to avoid them: employing a wider range of models, incorporating more realistic environmental conditions, better aligning studies between animals and patients, and focusing on human biology and therapeutic goals. Such changes are costly; but insisting it would be impractical or unrealistic to change strategies offers no way out of the current impasse. Rather, we must acknowledge the obstacles as well as the advantages presented by core models, and direct some of our investments in translational research toward getting around them.
Collapse
Affiliation(s)
- Jessica A Bolker
- Department of Biological Sciences, University of New Hampshire, Durham, NH, USA
| |
Collapse
|
221
|
Steven S, Dib M, Roohani S, Kashani F, Münzel T, Daiber A. Time Response of Oxidative/Nitrosative Stress and Inflammation in LPS-Induced Endotoxaemia-A Comparative Study of Mice and Rats. Int J Mol Sci 2017; 18:ijms18102176. [PMID: 29057830 PMCID: PMC5666857 DOI: 10.3390/ijms18102176] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 10/01/2017] [Accepted: 10/09/2017] [Indexed: 12/22/2022] Open
Abstract
Sepsis is a severe and multifactorial disease with a high mortality rate. It represents a strong inflammatory response to an infection and is associated with vascular inflammation and oxidative/nitrosative stress. Here, we studied the underlying time responses in the widely used lipopolysaccharide (LPS)-induced endotoxaemia model in mice and rats. LPS (10 mg/kg; from Salmonella Typhosa) was intraperitoneally injected into mice and rats. Animals of every species were divided into five groups and sacrificed at specific points in time (0, 3, 6, 9, 12 h). White blood cells (WBC) decreased significantly in both species after 3 h and partially recovered with time, whereas platelet decrease did not recover. Oxidative burst and iNOS-derived nitrosyl-iron hemoglobin (HbNO) increased with time (maxima at 9 or 12 h). Immune cell infiltration (CD68 and F4/80 content) showed an increase with time, which was supported by increased vascular mRNA expression of VCAM-1, P-selectin, IL-6 and TNF-α. We characterized the time responses of vascular inflammation and oxidative/nitrosative stress in LPS-induced endotoxaemic mice and rats. The results of this study will help to interpret and compare data from different animal species in LPS-induced endotoxaemia models for the identification of new drug targets.
Collapse
Affiliation(s)
- Sebastian Steven
- Center for Cardiology, Cardiology I, University Medical Center of the Johannes Gutenberg-University, D-55131 Mainz, Germany.
- Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University, D-55131 Mainz, Germany.
| | - Mobin Dib
- Center for Cardiology, Cardiology I, University Medical Center of the Johannes Gutenberg-University, D-55131 Mainz, Germany.
| | - Siyer Roohani
- Center for Cardiology, Cardiology I, University Medical Center of the Johannes Gutenberg-University, D-55131 Mainz, Germany.
| | - Fatemeh Kashani
- Center for Cardiology, Cardiology I, University Medical Center of the Johannes Gutenberg-University, D-55131 Mainz, Germany.
| | - Thomas Münzel
- Center for Cardiology, Cardiology I, University Medical Center of the Johannes Gutenberg-University, D-55131 Mainz, Germany.
| | - Andreas Daiber
- Center for Cardiology, Cardiology I, University Medical Center of the Johannes Gutenberg-University, D-55131 Mainz, Germany.
- Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University, D-55131 Mainz, Germany.
| |
Collapse
|
222
|
Bolton JL, Wiley MG, Ryan B, Truong S, Strait M, Baker DC, Yang NY, Ilkayeva O, O'Connell TM, Wroth SW, Sánchez CL, Swamy G, Newgard C, Kuhn C, Bilbo SD, Simmons LA. Perinatal western-type diet and associated gestational weight gain alter postpartum maternal mood. Brain Behav 2017; 7:e00828. [PMID: 29075574 PMCID: PMC5651398 DOI: 10.1002/brb3.828] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Revised: 07/07/2017] [Accepted: 08/06/2017] [Indexed: 12/11/2022] Open
Abstract
INTRODUCTION The role of perinatal diet in postpartum maternal mood disorders, including depression and anxiety, remains unclear. We investigated whether perinatal consumption of a Western-type diet (high in fat and branched-chain amino acids [BCAA]) and associated gestational weight gain (GWG) cause serotonin dysregulation in the central nervous system (CNS), resulting in postpartum depression and anxiety (PPD/A). METHODS Mouse dams were fed one of four diets (high-fat/high BCAA, low-fat/high BCAA, high-fat, and low-fat) prior to mating and throughout gestation and lactation. Postpartum behavioral assessments were conducted, and plasma and brain tissues assayed. To evaluate potential clinical utility, we conducted preliminary human studies using data from an extant sample of 17 primiparous women with high GWG, comparing across self-reported postpartum mood symptoms using the Edinburgh Postnatal Depression Scale (EPDS) for percent GWG and plasma amino acid levels. RESULTS Mouse dams fed the high-fat/high BCAA diet gained more weight per kcal consumed, and BCAA-supplemented dams lost weight more slowly postpartum. Dams on BCAA-supplemented diets exhibited increased PPD/A-like behavior, decreased dopaminergic function, and decreased plasma tyrosine and histidine levels when assessed on postnatal day (P)8. Preliminary human data showed that GWG accounted for 29% of the variance in EPDS scores. Histidine was also lower in women with higher EPDS scores. CONCLUSIONS These findings highlight the role of perinatal diet and excess GWG in the development of postpartum mood disorders.
Collapse
Affiliation(s)
- Jessica L Bolton
- Department of Psychology and Neuroscience Duke University Durham NC USA
| | - Melanie G Wiley
- Department of Psychology and Neuroscience Duke University Durham NC USA
| | - Bailey Ryan
- Department of Psychology and Neuroscience Duke University Durham NC USA
| | - Samantha Truong
- Department of Psychology and Neuroscience Duke University Durham NC USA
| | | | | | | | - Olga Ilkayeva
- Duke Molecular Physiology Institute Duke University School of Medicine Durham NC USA
| | - Thomas M O'Connell
- Duke Molecular Physiology Institute Duke University School of Medicine Durham NC USA
| | | | - Cristina L Sánchez
- Department of Pharmacology and Cancer Biology Duke University School of Medicine Durham NC USA
| | - Geeta Swamy
- Department of Obstetrics and Gynecology Duke University School of Medicine Durham NC USA
| | - Christopher Newgard
- Duke Molecular Physiology Institute Duke University School of Medicine Durham NC USA.,Department of Pharmacology and Cancer Biology Duke University School of Medicine Durham NC USA
| | - Cynthia Kuhn
- Department of Pharmacology and Cancer Biology Duke University School of Medicine Durham NC USA
| | - Staci D Bilbo
- Department of Psychology and Neuroscience Duke University Durham NC USA
| | | |
Collapse
|
223
|
Translational Aspects of Diet and Non-Alcoholic Fatty Liver Disease. Nutrients 2017; 9:nu9101077. [PMID: 28956824 PMCID: PMC5691694 DOI: 10.3390/nu9101077] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 09/25/2017] [Accepted: 09/26/2017] [Indexed: 12/12/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a spectrum of diseases ranging from simple steatosis without inflammation or fibrosis to nonalcoholic steatohepatitis (NASH). Despite the strong association between dietary factors and NAFLD, no dietary animal model of NAFLD fully recapitulates the complex metabolic and histological phenotype of the disease, although recent models show promise. Although animal models have significantly contributed to our understanding of human diseases, they have been less successful in accurate translation to predict effective treatment strategies. We discuss strategies to overcome this challenge, in particular the adoption of big data approaches combining clinical phenotype, genomic heterogeneity, transcriptomics, and metabolomics changes to identify the ideal NAFLD animal model for a given scientific question or to test a given drug. We conclude by noting that novel big data approaches may help to bridge the translational gap for selecting dietary models of NAFLD.
Collapse
|
224
|
Highly Variable Genomic Landscape of Endogenous Retroviruses in the C57BL/6J Inbred Strain, Depending on Individual Mouse, Gender, Organ Type, and Organ Location. Int J Genomics 2017; 2017:3152410. [PMID: 28951865 PMCID: PMC5603323 DOI: 10.1155/2017/3152410] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 06/16/2017] [Accepted: 07/03/2017] [Indexed: 11/17/2022] Open
Abstract
Transposable repetitive elements, named the "TREome," represent ~40% of the mouse genome. We postulate that the germ line genome undergoes temporal and spatial diversification into somatic genomes in conjunction with the TREome activity. C57BL/6J inbred mice were subjected to genomic landscape analyses using a TREome probe from murine leukemia virus-type endogenous retroviruses (MLV-ERVs). None shared the same MLV-ERV landscape within each comparison group: (1) sperm and 18 tissues from one mouse, (2) six brain compartments from two females, (3) spleen and thymus samples from four age groups, (4) three spatial tissue sets from two females, and (5) kidney and liver samples from three females and three males. Interestingly, males had more genomic MLV-ERV copies than females; moreover, only in the males, the kidneys had higher MLV-ERV copies than the livers. Perhaps, the mouse-, gender-, and tissue/cell-dependent MLV-ERV landscapes are linked to the individual-specific and dynamic phenotypes of the C57BL/6J inbred population.
Collapse
|
225
|
Influenza Pathogenesis in Genetically Defined Resistant and Susceptible Murine Strains
. THE YALE JOURNAL OF BIOLOGY AND MEDICINE 2017; 90:471-479. [PMID: 28955185 PMCID: PMC5612189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
The murine infection model is a cornerstone for influenza virus research and includes aspects such as disease pathogenesis, immunobiology, and vaccine and antiviral drug development. One compelling feature of the murine model is the availability of inbred mouse strains, each with a unique genetic makeup and potential for variable responses to influenza infection. Using highly controlled infection studies, the response to influenza virus infection is classified on a spectrum from susceptible to resistant, reflecting severe morbidity and high mortality, to limited or no morbidity and no mortality. Although there have been a variety of studies establishing disparate pathogenesis amongst various murine strains, thus far, there is no consensus regarding the determinants of the outcome of infection. The goal of this review is to explore and discuss the differences in pathogenesis, as well as the innate and adaptive immune responses to influenza infection that have been described in susceptible and resistant mouse strains. Understanding how host genetics influences the response to influenza infection provides valuable insight into the variable responses seen in vaccine or drug efficacy studies, as well as indicates possible mechanisms contributing to increased disease severity in humans infected with influenza virus with no known risk factors.
Collapse
|
226
|
Liu J, Kumar S, Dolzhenko E, Alvarado GF, Guo J, Lu C, Chen Y, Li M, Dessing MC, Parvez RK, Cippà PE, Krautzberger AM, Saribekyan G, Smith AD, McMahon AP. Molecular characterization of the transition from acute to chronic kidney injury following ischemia/reperfusion. JCI Insight 2017; 2:94716. [PMID: 28931758 PMCID: PMC5612583 DOI: 10.1172/jci.insight.94716] [Citation(s) in RCA: 215] [Impact Index Per Article: 26.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Accepted: 08/10/2017] [Indexed: 12/16/2022] Open
Abstract
Though an acute kidney injury (AKI) episode is associated with an increased risk of chronic kidney disease (CKD), the mechanisms determining the transition from acute to irreversible chronic injury are not well understood. To extend our understanding of renal repair, and its limits, we performed a detailed molecular characterization of a murine ischemia/reperfusion injury (IRI) model for 12 months after injury. Together, the data comprising RNA-sequencing (RNA-seq) analysis at multiple time points, histological studies, and molecular and cellular characterization of targeted gene activity provide a comprehensive profile of injury, repair, and long-term maladaptive responses following IRI. Tubular atrophy, interstitial fibrosis, inflammation, and development of multiple renal cysts were major long-term outcomes of IRI. Progressive proximal tubular injury tracks with de novo activation of multiple Krt genes, including Krt20, a biomarker of renal tubule injury. RNA-seq analysis highlights a cascade of temporal-specific gene expression patterns related to tubular injury/repair, fibrosis, and innate and adaptive immunity. Intersection of these data with human kidney transplant expression profiles identified overlapping gene expression signatures correlating with different stages of the murine IRI response. The comprehensive characterization of incomplete recovery after ischemic AKI provides a valuable resource for determining the underlying pathophysiology of human CKD.
Collapse
Affiliation(s)
- Jing Liu
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| | - Sanjeev Kumar
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA.,Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Egor Dolzhenko
- Molecular and Computational Biology, Division of Biological Sciences, University of Southern California, Los Angeles, California, USA
| | - Gregory F Alvarado
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| | - Jinjin Guo
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| | - Can Lu
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| | - Yibu Chen
- Norris Medical Library, University of Southern California, Los Angeles, California
| | - Meng Li
- Norris Medical Library, University of Southern California, Los Angeles, California
| | - Mark C Dessing
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| | - Riana K Parvez
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| | - Pietro E Cippà
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| | - A Michaela Krautzberger
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| | - Gohar Saribekyan
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| | - Andrew D Smith
- Molecular and Computational Biology, Division of Biological Sciences, University of Southern California, Los Angeles, California, USA
| | - Andrew P McMahon
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| |
Collapse
|
227
|
Domaszewska T, Scheuermann L, Hahnke K, Mollenkopf H, Dorhoi A, Kaufmann SHE, Weiner J. Concordant and discordant gene expression patterns in mouse strains identify best-fit animal model for human tuberculosis. Sci Rep 2017; 7:12094. [PMID: 28935874 PMCID: PMC5608750 DOI: 10.1038/s41598-017-11812-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Accepted: 08/30/2017] [Indexed: 12/22/2022] Open
Abstract
Immunity in infection, inflammation and malignancy differs markedly in man and mouse. Still, we learn about human immunity in large extent from experimental mouse models. We propose a novel data integration approach which identifies concordant and discordant gene expression patterns of the immune responses in heterologous data sets. We have conducted experiments to compare human and murine transcriptional responses to Mycobacterium tuberculosis (Mtb) infection in whole blood (WB) as well as macrophages and compared them with simulated as well as publicly available data. Our results indicate profound differences between patterns of gene expression in innate and adaptive immunity in man and mouse upon Mtb infection. We characterized differential expression of T-cell related genes corresponding to the differences in phenotype between tuberculosis (TB) highly and low susceptible mouse strains. Our approach is general and facilitates the choice of optimal animal model for studies of the human immune response to a particular disease.
Collapse
Affiliation(s)
- Teresa Domaszewska
- Max Planck Institute for Infection Biology, Department of Immunology, Charitéplatz 1, D-10117, Berlin, Germany
| | - Lisa Scheuermann
- Max Planck Institute for Infection Biology, Department of Immunology, Charitéplatz 1, D-10117, Berlin, Germany
| | - Karin Hahnke
- Max Planck Institute for Infection Biology, Department of Immunology, Charitéplatz 1, D-10117, Berlin, Germany
| | - Hans Mollenkopf
- Max Planck Institute for Infection Biology, Department of Immunology, Charitéplatz 1, D-10117, Berlin, Germany
| | - Anca Dorhoi
- Max Planck Institute for Infection Biology, Department of Immunology, Charitéplatz 1, D-10117, Berlin, Germany
| | - Stefan H E Kaufmann
- Max Planck Institute for Infection Biology, Department of Immunology, Charitéplatz 1, D-10117, Berlin, Germany.
| | - January Weiner
- Max Planck Institute for Infection Biology, Department of Immunology, Charitéplatz 1, D-10117, Berlin, Germany.
| |
Collapse
|
228
|
Devarapu SK, Grill JF, Xie J, Weidenbusch M, Honarpisheh M, Vielhauer V, Anders HJ, Mulay SR. Tumor necrosis factor superfamily ligand mRNA expression profiles differ between humans and mice during homeostasis and between various murine kidney injuries. J Biomed Sci 2017; 24:77. [PMID: 28927419 PMCID: PMC5606058 DOI: 10.1186/s12929-017-0383-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 09/14/2017] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Several tumour necrosis factor (TNF) based therapeutics have already been approved for human use and several others are emerging. Therefore, we determined the mRNA expression levels of the TNF superfamily ligands (TNFSF) - e.g. TNF-α, lymphotoxin (LT)-α, LT-β, Fas-L (CD95-L), TNF-related apoptosis-inducing ligand (TRAIL), TNF-related weak inducer of apoptosis (TWEAK), 4-1BBL, OX40-L (CD252) and amyloid precursor protein (APP) in healthy human and mouse solid organs. METHODS We used quantitative real time-PCR to analyse mRNA expression levels of TNFSF ligands. Murine models of acute ischemic renal injury, chronic oxalate nephropathy, and immune complex glomerulonephritis were used. Renal injury was assessed by PAS staining, and infiltrating immune cells were analysed by immunohistochemistry. Data was analysed using non-parametric ANOVA (non-parametric; Kruskal-Wallis test). RESULTS We observed significant differences in the mRNA expression levels of TNFSF ligands in human and mouse solid organs. Furthermore, we determined their mRNA expressions during acute and chronic kidney injuries in mice. Our data demonstrate that the mRNA expression levels of TNFSF vary depending on the type of tissue injury - for example, acute ischemic renal injury, chronic crystalline nephropathy, and immune complex glomerulonephritis. In addition, we observed that mRNA expressions of TNFSF ligands are differentially regulated during the course of a transient ischemic renal injury (IRI) and chronic kidney modelling. We observed that TNF-α, LT-β, and 4-1BBL were significantly upregulated during the progression of IRI and crystal-induced chronic kidney disease (CKD), whereas only 4-1BBL and TNF-α were significantly upregulated and LT-β was significantly downregulated during the progression of immune complex glomerulonephritis. The mRNA expression of Fas-L was higher during IRI whereas it decreased in a time dependent manner during the progression of crystal-induced CKD. CONCLUSION We conclude that the injury- and species-specific differences of TNFSF ligands must be considered in order to avoid the misinterpretation and wrong conclusions during data extrapolation between species.
Collapse
Affiliation(s)
- Satish Kumar Devarapu
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Munich, Germany
| | - Julia Felicitas Grill
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Munich, Germany
| | - Junhui Xie
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Munich, Germany.,Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong, University of Science and Technology, Wuhan, China
| | - Marc Weidenbusch
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Munich, Germany
| | - Mohsen Honarpisheh
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Munich, Germany
| | - Volker Vielhauer
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Munich, Germany
| | - Hans-Joachim Anders
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Munich, Germany
| | - Shrikant R Mulay
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Munich, Germany. .,Nephrologisches Zentrum, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Schillerstr. 42, D-80336, Munich, Germany.
| |
Collapse
|
229
|
Burgdorf T, Dunst S, Ertych N, Fetz V, Violet N, Vogl S, Schönfelder G, Schwarz F, Oelgeschläger M. The AOP Concept: How Novel Technologies Can Support Development of Adverse Outcome Pathways. ACTA ACUST UNITED AC 2017. [DOI: 10.1089/aivt.2017.0011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Tanja Burgdorf
- Department Experimental Toxicology and ZEBET, German Centre for The Protection of Laboratory Animals (Bf3R), German Federal Institute for Risk Assessment, Berlin, Germany
| | - Sebastian Dunst
- Department Experimental Toxicology and ZEBET, German Centre for The Protection of Laboratory Animals (Bf3R), German Federal Institute for Risk Assessment, Berlin, Germany
| | - Norman Ertych
- Department Experimental Toxicology and ZEBET, German Centre for The Protection of Laboratory Animals (Bf3R), German Federal Institute for Risk Assessment, Berlin, Germany
| | - Verena Fetz
- Department Experimental Toxicology and ZEBET, German Centre for The Protection of Laboratory Animals (Bf3R), German Federal Institute for Risk Assessment, Berlin, Germany
| | - Norman Violet
- Department Experimental Toxicology and ZEBET, German Centre for The Protection of Laboratory Animals (Bf3R), German Federal Institute for Risk Assessment, Berlin, Germany
| | - Silvia Vogl
- Department Experimental Toxicology and ZEBET, German Centre for The Protection of Laboratory Animals (Bf3R), German Federal Institute for Risk Assessment, Berlin, Germany
| | - Gilbert Schönfelder
- Department Experimental Toxicology and ZEBET, German Centre for The Protection of Laboratory Animals (Bf3R), German Federal Institute for Risk Assessment, Berlin, Germany
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Clinical Pharmacology and Toxicology, Berlin, Germany
| | - Franziska Schwarz
- Department Experimental Toxicology and ZEBET, German Centre for The Protection of Laboratory Animals (Bf3R), German Federal Institute for Risk Assessment, Berlin, Germany
| | - Michael Oelgeschläger
- Department Experimental Toxicology and ZEBET, German Centre for The Protection of Laboratory Animals (Bf3R), German Federal Institute for Risk Assessment, Berlin, Germany
| |
Collapse
|
230
|
Lam D, Lively S, Schlichter LC. Responses of rat and mouse primary microglia to pro- and anti-inflammatory stimuli: molecular profiles, K + channels and migration. J Neuroinflammation 2017; 14:166. [PMID: 28830445 PMCID: PMC5567442 DOI: 10.1186/s12974-017-0941-3] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 08/13/2017] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Acute CNS damage is commonly studied using rat and mouse models, but increasingly, molecular analysis is finding species differences that might affect the ability to translate findings to humans. Microglia can undergo complex molecular and functional changes, often studied by in vitro responses to discrete activating stimuli. There is considerable evidence that pro-inflammatory (M1) activation can exacerbate tissue damage, while anti-inflammatory (M2) states help resolve inflammation and promote tissue repair. However, in assessing potential therapeutic targets for controlling inflammation, it is crucial to determine whether rat and mouse microglia respond the same. METHODS Primary microglia from Sprague-Dawley rats and C57BL/6 mice were cultured, then stimulated with interferon-γ + tumor necrosis factor-α (I + T; M1 activation), interleukin (IL)-4 (M2a, alternative activation), or IL-10 (M2c, acquired deactivation). To profile their activation responses, NanoString was used to monitor messenger RNA (mRNA) expression of numerous pro- and anti-inflammatory mediators, microglial markers, immunomodulators, and other molecules. Western analysis was used to measure selected proteins. Two potential targets for controlling inflammation-inward- and outward-rectifier K+ channels (Kir2.1, Kv1.3)-were examined (mRNA, currents) and specific channel blockers were applied to determine their contributions to microglial migration in the different activation states. RESULTS Pro-inflammatory molecules increased after I + T treatment but there were several qualitative and quantitative differences between the species (e.g., iNOS and nitric oxide, COX-2). Several molecules commonly associated with an M2a state differed between species or they were induced in additional activation states (e.g., CD206, ARG1). Resting levels and/or responses of several microglial markers (Iba1, CD11b, CD68) differed with the activation state, species, or both. Transcripts for several Kir2 and Kv1 family members were detected in both species. However, the current amplitudes (mainly Kir2.1 and Kv1.3) depended on activation state and species. Treatment-induced changes in morphology and migratory capacity were similar between the species (migration reduced by I + T, increased by IL-4 or IL-10). In both species, Kir2.1 block reduced migration and Kv1.3 block increased it, regardless of activation state; thus, these channels might affect microglial migration to damage sites. CONCLUSIONS Caution is recommended in generalizing molecular and functional responses of microglia to activating stimuli between species.
Collapse
Affiliation(s)
- Doris Lam
- Genes and Development Division, Krembil Research Institute, University Health Network, Krembil Discovery Tower, Room 7KD417, 60 Leonard Avenue, Toronto, ON, M5T 2S8, Canada.,Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - Starlee Lively
- Genes and Development Division, Krembil Research Institute, University Health Network, Krembil Discovery Tower, Room 7KD417, 60 Leonard Avenue, Toronto, ON, M5T 2S8, Canada
| | - Lyanne C Schlichter
- Genes and Development Division, Krembil Research Institute, University Health Network, Krembil Discovery Tower, Room 7KD417, 60 Leonard Avenue, Toronto, ON, M5T 2S8, Canada. .,Department of Physiology, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
231
|
Sweeney TE, Lofgren S, Khatri P, Rogers AJ. Gene Expression Analysis to Assess the Relevance of Rodent Models to Human Lung Injury. Am J Respir Cell Mol Biol 2017; 57:184-192. [PMID: 28324666 DOI: 10.1165/rcmb.2016-0395oc] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The relevance of animal models to human diseases is an area of intense scientific debate. The degree to which mouse models of lung injury recapitulate human lung injury has never been assessed. Integrating data from both human and animal expression studies allows for increased statistical power and identification of conserved differential gene expression across organisms and conditions. We sought comprehensive integration of gene expression data in experimental acute lung injury (ALI) in rodents compared with humans. We performed two separate gene expression multicohort analyses to determine differential gene expression in experimental animal and human lung injury. We used correlational and pathway analyses combined with external in vitro gene expression data to identify both potential drivers of underlying inflammation and therapeutic drug candidates. We identified 21 animal lung tissue datasets and three human lung injury bronchoalveolar lavage datasets. We show that the metasignatures of animal and human experimental ALI are significantly correlated despite these widely varying experimental conditions. The gene expression changes among mice and rats across diverse injury models (ozone, ventilator-induced lung injury, LPS) are significantly correlated with human models of lung injury (Pearson r = 0.33-0.45, P < 1E-16). Neutrophil signatures are enriched in both animal and human lung injury. Predicted therapeutic targets, peptide ligand signatures, and pathway analyses are also all highly overlapping. Gene expression changes are similar in animal and human experimental ALI, and provide several physiologic and therapeutic insights to the disease.
Collapse
Affiliation(s)
- Timothy E Sweeney
- 1 Stanford Institute for Immunity, Transplantation and Infection.,2 Biomedical Informatics Research, and
| | - Shane Lofgren
- 1 Stanford Institute for Immunity, Transplantation and Infection.,2 Biomedical Informatics Research, and
| | - Purvesh Khatri
- 1 Stanford Institute for Immunity, Transplantation and Infection.,2 Biomedical Informatics Research, and
| | - Angela J Rogers
- 3 Department of Medicine, Division of Pulmonary and Critical Care Medicine, Stanford University School of Medicine, Stanford, California
| |
Collapse
|
232
|
Weidner C, Steinfath M, Wistorf E, Oelgeschläger M, Schneider MR, Schönfelder G. A Protocol for Using Gene Set Enrichment Analysis to Identify the Appropriate Animal Model for Translational Research. J Vis Exp 2017. [PMID: 28872130 DOI: 10.3791/55768] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Recent studies that compared transcriptomic datasets of human diseases with datasets from mouse models using traditional gene-to-gene comparison techniques resulted in contradictory conclusions regarding the relevance of animal models for translational research. A major reason for the discrepancies between different gene expression analyses is the arbitrary filtering of differentially expressed genes. Furthermore, the comparison of single genes between different species and platforms often is limited by technical variance, leading to misinterpretation of the con/discordance between data from human and animal models. Thus, standardized approaches for systematic data analysis are needed. To overcome subjective gene filtering and ineffective gene-to-gene comparisons, we recently demonstrated that gene set enrichment analysis (GSEA) has the potential to avoid these problems. Therefore, we developed a standardized protocol for the use of GSEA to distinguish between appropriate and inappropriate animal models for translational research. This protocol is not suitable to predict how to design new model systems a-priori, as it requires existing experimental omics data. However, the protocol describes how to interpret existing data in a standardized manner in order to select the most suitable animal model, thus avoiding unnecessary animal experiments and misleading translational studies.
Collapse
Affiliation(s)
- Christopher Weidner
- Department of Experimental Toxicology and ZEBET, German Federal Institute for Risk Assessment (BfR)
| | - Matthias Steinfath
- Department of Experimental Toxicology and ZEBET, German Federal Institute for Risk Assessment (BfR)
| | - Elisa Wistorf
- Department of Experimental Toxicology and ZEBET, German Federal Institute for Risk Assessment (BfR)
| | - Michael Oelgeschläger
- Department of Experimental Toxicology and ZEBET, German Federal Institute for Risk Assessment (BfR)
| | - Marlon R Schneider
- Department of Experimental Toxicology and ZEBET, German Federal Institute for Risk Assessment (BfR)
| | - Gilbert Schönfelder
- Department of Experimental Toxicology and ZEBET, German Federal Institute for Risk Assessment (BfR); Department of Clinical Pharmacology and Toxicology, Charité-Universitätsmedizin Berlin;
| |
Collapse
|
233
|
Abstract
Humans prefer to live within their thermal comfort or neutral zone, which they create by making shelters, wearing clothing and, more recently, by regulating their ambient temperature. These strategies enable humans to maintain a constant core temperature (a trait that is conserved across all endotherms, including mammals and birds) with minimal energy expenditure. Although this primordial drive leads us to seek thermal comfort, we house our experimental animals, laboratory mice (Mus musculus), under conditions of thermal stress. In this Review, we discuss how housing mice below their thermoneutral zone limits our ability to model and study human diseases. Using examples from cardiovascular physiology, metabolic disorders, infections and tumour immunology, we show that certain phenotypes observed under conditions of thermal stress disappear when mice are housed at thermoneutrality, whereas others emerge that are more consistent with human biology. Thus, we propose that warming the mouse might enable more predictive modelling of human diseases and therapies.
Collapse
Affiliation(s)
- Kirthana Ganeshan
- Cardiovascular Research Institute, University of California, San Francisco, 555 Mission Bay Boulevard South, San Francisco, California 94158, USA
| | - Ajay Chawla
- Cardiovascular Research Institute, University of California, San Francisco, 555 Mission Bay Boulevard South, San Francisco, California 94158, USA
- Department of Physiology University of California, San Francisco
- Department of Medicine, University of California, San Francisco, 505 Parnassus Avenue, San Francisco, California 94143, USA
| |
Collapse
|
234
|
Bron AJ, de Paiva CS, Chauhan SK, Bonini S, Gabison EE, Jain S, Knop E, Markoulli M, Ogawa Y, Perez V, Uchino Y, Yokoi N, Zoukhri D, Sullivan DA. TFOS DEWS II pathophysiology report. Ocul Surf 2017; 15:438-510. [PMID: 28736340 DOI: 10.1016/j.jtos.2017.05.011] [Citation(s) in RCA: 1139] [Impact Index Per Article: 142.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 05/26/2017] [Indexed: 12/18/2022]
Abstract
The TFOS DEWS II Pathophysiology Subcommittee reviewed the mechanisms involved in the initiation and perpetuation of dry eye disease. Its central mechanism is evaporative water loss leading to hyperosmolar tissue damage. Research in human disease and in animal models has shown that this, either directly or by inducing inflammation, causes a loss of both epithelial and goblet cells. The consequent decrease in surface wettability leads to early tear film breakup and amplifies hyperosmolarity via a Vicious Circle. Pain in dry eye is caused by tear hyperosmolarity, loss of lubrication, inflammatory mediators and neurosensory factors, while visual symptoms arise from tear and ocular surface irregularity. Increased friction targets damage to the lids and ocular surface, resulting in characteristic punctate epithelial keratitis, superior limbic keratoconjunctivitis, filamentary keratitis, lid parallel conjunctival folds, and lid wiper epitheliopathy. Hybrid dry eye disease, with features of both aqueous deficiency and increased evaporation, is common and efforts should be made to determine the relative contribution of each form to the total picture. To this end, practical methods are needed to measure tear evaporation in the clinic, and similarly, methods are needed to measure osmolarity at the tissue level across the ocular surface, to better determine the severity of dry eye. Areas for future research include the role of genetic mechanisms in non-Sjögren syndrome dry eye, the targeting of the terminal duct in meibomian gland disease and the influence of gaze dynamics and the closed eye state on tear stability and ocular surface inflammation.
Collapse
Affiliation(s)
- Anthony J Bron
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK; Vision and Eye Research Unit, Anglia Ruskin University, Cambridge, UK.
| | - Cintia S de Paiva
- Department of Ophthalmology, Baylor College of Medicine, Houston, TX, USA
| | - Sunil K Chauhan
- Schepens Eye Research Institute & Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Stefano Bonini
- Department of Ophthalmology, University Campus Biomedico, Rome, Italy
| | - Eric E Gabison
- Department of Ophthalmology, Fondation Ophtalmologique Rothschild & Hôpital Bichat Claude Bernard, Paris, France
| | - Sandeep Jain
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL, USA
| | - Erich Knop
- Departments of Cell and Neurobiology and Ocular Surface Center Berlin, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Maria Markoulli
- School of Optometry and Vision Science, University of New South Wales, Sydney, Australia
| | - Yoko Ogawa
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
| | - Victor Perez
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami, Miami, FL, USA
| | - Yuichi Uchino
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
| | - Norihiko Yokoi
- Department of Ophthalmology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Driss Zoukhri
- Tufts University School of Dental Medicine, Boston, MA, USA
| | - David A Sullivan
- Schepens Eye Research Institute & Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
235
|
Perry CJ, Lawrence AJ. Hurdles in Basic Science Translation. Front Pharmacol 2017; 8:478. [PMID: 28769807 PMCID: PMC5513913 DOI: 10.3389/fphar.2017.00478] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 07/03/2017] [Indexed: 11/13/2022] Open
Abstract
In the past century there have been incredible advances in the field of medical research, but what hinders translation of this knowledge into effective treatment for human disease? There is an increasing focus on the failure of many research breakthroughs to be translated through the clinical trial process and into medical practice. In this mini review, we will consider some of the reasons that findings in basic medical research fail to become translated through clinical trials and into basic medical practices. We focus in particular on the way that human disease is modeled, the understanding we have of how our targets behave in vivo, and also some of the issues surrounding reproducibility of basic research findings. We will also look at some of the ways that have been proposed for overcoming these issues. It appears that there needs to be a cultural shift in the way we fund, publish and recognize quality control in scientific research. Although this is a daunting proposition, we hope that with increasing awareness and focus on research translation and the hurdles that impede it, the field of medical research will continue to inform and improve medical practice across the world.
Collapse
Affiliation(s)
- Christina J Perry
- Behavioural Neuroscience Division, The Florey Institute of Neuroscience and Mental Health, ParkvilleVIC, Australia.,Florey Department of Neuroscience and Mental Health, University of Melbourne, MelbourneVIC, Australia
| | - Andrew J Lawrence
- Behavioural Neuroscience Division, The Florey Institute of Neuroscience and Mental Health, ParkvilleVIC, Australia.,Florey Department of Neuroscience and Mental Health, University of Melbourne, MelbourneVIC, Australia
| |
Collapse
|
236
|
Masopust D, Sivula CP, Jameson SC. Of Mice, Dirty Mice, and Men: Using Mice To Understand Human Immunology. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2017; 199:383-388. [PMID: 28696328 PMCID: PMC5512602 DOI: 10.4049/jimmunol.1700453] [Citation(s) in RCA: 208] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Accepted: 04/28/2017] [Indexed: 12/29/2022]
Abstract
Mouse models have enabled breakthroughs in our understanding of the immune system, but it has become increasingly popular to emphasize their shortcomings when translating observations to humans. This review provides a brief summary of mouse natural history, husbandry, and the pros and cons of pursuing basic research in mice versus humans. Opportunities are discussed for extending the predictive translational value of mouse research, with an emphasis on exploitation of a "dirty" mouse model that better mimics the diverse infectious history that is typical of most humans.
Collapse
Affiliation(s)
- David Masopust
- Department of Microbiology and Immunology, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455;
| | - Christine P Sivula
- Research Animal Resources, University of Minnesota, Minneapolis, MN 55455; and
| | - Stephen C Jameson
- Department of Laboratory Medicine and Pathology, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455
| |
Collapse
|
237
|
Ntougkos E, Chouvardas P, Roumelioti F, Ospelt C, Frank-Bertoncelj M, Filer A, Buckley CD, Gay S, Nikolaou C, Kollias G. Genomic Responses of Mouse Synovial Fibroblasts During Tumor Necrosis Factor-Driven Arthritogenesis Greatly Mimic Those in Human Rheumatoid Arthritis. Arthritis Rheumatol 2017; 69:1588-1600. [PMID: 28409894 DOI: 10.1002/art.40128] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 04/11/2017] [Indexed: 12/30/2022]
Abstract
OBJECTIVE Aberrant activation of synovial fibroblasts is a key determinant in the pathogenesis of rheumatoid arthritis (RA). The aims of this study were to produce a map of gene expression and epigenetic changes occurring in this cell type during disease progression in the human tumor necrosis factor (TNF)-transgenic model of arthritis and to identify commonalities with human synovial fibroblasts. METHODS We used deep sequencing to probe the transcriptome, the methylome, and the chromatin landscape of cultured mouse arthritogenic synovial fibroblasts at 3 stages of disease, as well as synovial fibroblasts stimulated with human TNF. We performed bioinformatics analyses at the gene, pathway, and network levels, compared mouse and human data, and validated selected genes in both species. RESULTS We found that synovial fibroblast arthritogenicity was reflected in distinct dynamic patterns of transcriptional dysregulation, which was especially enriched in pathways of the innate immune response and mesenchymal differentiation. A functionally representative subset of these changes was associated with methylation, mostly in gene bodies. The arthritogenic state involved highly active promoters, which were marked by histone H3K4 trimethylation. There was significant overlap between the mouse and human data at the level of dysregulated genes and to an even greater extent at the level of pathways. CONCLUSION This study is the first systematic examination of the pathogenic changes that occur in mouse synovial fibroblasts during progressive TNF-driven arthritogenesis. Significant correlations with the respective human RA synovial fibroblast data further validate the human TNF-transgenic mouse as a reliable model of the human disease. The resource of data generated in this work may serve as a framework for the discovery of novel pathogenic mechanisms and disease biomarkers.
Collapse
Affiliation(s)
| | - Panagiotis Chouvardas
- BSRC Alexander Fleming, Vari, Greece, and National and Kapodistrian University of Athens, Athens, Greece
| | - Fani Roumelioti
- BSRC Alexander Fleming, Vari, Greece, and National and Kapodistrian University of Athens, Athens, Greece
| | | | | | | | | | - Steffen Gay
- University Hospital of Zurich, Zurich, Switzerland
| | | | - George Kollias
- BSRC Alexander Fleming, Vari, Greece, and National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
238
|
Abstract
Cross-species comparisons of genomes, transcriptomes and gene regulation are now feasible at unprecedented resolution and throughput, enabling the comparison of human and mouse biology at the molecular level. Insights have been gained into the degree of conservation between human and mouse at the level of not only gene expression but also epigenetics and inter-individual variation. However, a number of limitations exist, including incomplete transcriptome characterization and difficulties in identifying orthologous phenotypes and cell types, which are beginning to be addressed by emerging technologies. Ultimately, these comparisons will help to identify the conditions under which the mouse is a suitable model of human physiology and disease, and optimize the use of animal models.
Collapse
Affiliation(s)
- Alessandra Breschi
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr. Aiguader 88, 08003 Barcelona, Spain
- Universitat Pompeu Fabra (UPF), 08002 Barcelona, Spain
| | - Thomas R Gingeras
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11742, USA
| | - Roderic Guigó
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr. Aiguader 88, 08003 Barcelona, Spain
- Universitat Pompeu Fabra (UPF), 08002 Barcelona, Spain
| |
Collapse
|
239
|
Shusterman A, Munz M, Richter G, Jepsen S, Lieb W, Krone B, Hoffman P, Laudes M, Wellmann J, Berger K, Kocher T, Offenbacher S, Divaris K, Franke A, Schreiber S, Dommisch H, Weiss E, Schaefer A, Houri-Haddad Y, Iraqi F. The PF4/PPBP/CXCL5 Gene Cluster Is Associated with Periodontitis. J Dent Res 2017; 96:945-952. [PMID: 28467728 PMCID: PMC5502958 DOI: 10.1177/0022034517706311] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Periodontitis is a common dysbiotic inflammatory disease with an estimated heritability of 50%. Due to the limited sample size of available periodontitis cohorts and the underlying trait heterogeneity, genome-wide association studies (GWAS) of chronic periodontitis (CP) have been unsuccessful in discovering susceptibility factors. A strategy that combines agnostic GWAS with a well-powered candidate-gene approach has the potential to discover novel loci. We combined RNA-seq data from gingival tissues with quantitative trait loci (QTLs) that were identified in a F2-cross of mice resistant and susceptible to infection with oral bacterial pathogens. Four genes, which were located within the mapped QTLs, showed differential expression. The chromosomal regions across the human orthologous were interrogated for putative periodontitis-associated variants using existing GWAS data from a German case-control sample of aggressive periodontitis (AgP; 651 cases, 4,001 controls), the most severe and early onset form of periodontitis. Two haplotype blocks, one upstream to the coding region of UGT2A1 (rs146712414, P = 9.1 × 10-5; odds ratio [OR], 1.34; 95% confidence interval [CI], 1.16-1.56) and one downstream of the genes PF4/PPBP/CXCL5 (rs1595009, P = 1.3 × 10-4; OR, 1.32; 95% CI, 1.15-1.52), were associated with AgP. The association of rs1595009 was validated in an independent cohort of CP of European Americans (1,961 cases and 1,864 controls; P = 0.03; OR, 1.45; 95% CI, 1.01-1.29). This association was further replicated in another sample of 399 German CP cases (disease onset <60 y of age) and 1,633 controls ( P = 0.03; OR, 1.75; 95% CI, 1.06-2.90). The combined estimates of association from all samples were P = 2.9 × 10-5 (OR, 1.2; 95% CI, 1.1-1.3). This study shows the strength of combining QTL mapping and RNA-Seq data from a mouse model with association studies in human case-control samples to identify genetic risk variants of periodontitis.
Collapse
Affiliation(s)
- A. Shusterman
- Department of Prosthodontics, Hadassah Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel
| | - M. Munz
- Department of Periodontology and Synoptic Medicine, Institute for Dental and Craniofacial Sciences, Charité–University Medicine Berlin, Berlin, Germany
- Institute for Integrative and Experimental Genomics, University Medical Center Schleswig-Holstein–Campus Lübeck, Lübeck, Germany
| | - G. Richter
- Department of Periodontology and Synoptic Medicine, Institute for Dental and Craniofacial Sciences, Charité–University Medicine Berlin, Berlin, Germany
| | - S. Jepsen
- Department of Periodontology, Operative and Preventive Dentistry, University of Bonn, Bonn, Germany
| | - W. Lieb
- Institute of Epidemiology, Biobank popgen, Christian-Albrechts-University, Kiel, Germany
| | - B. Krone
- Institute of Medical Informatics, Biometry and Epidemiology, University Clinic Essen, Essen, Germany
| | - P. Hoffman
- Institute of Human Genetics, University of Bonn, Bonn, Germany
- Germany und Human Genomics Research Group, Department of Biomedicine, University Hospital of Basel, Basel, Switzerland
| | - M. Laudes
- Clinic of Internal Medicine, University Clinic Schleswig-Holstein, Kiel, Germany
| | - J. Wellmann
- Institute of Epidemiology and Social Medicine, University of Münster, Münster, Germany
| | - K. Berger
- Institute of Epidemiology and Social Medicine, University of Münster, Münster, Germany
| | - T. Kocher
- Unit of Periodontology, Department of Restorative Dentistry, Periodontology, Endodontology, Preventive Dentistry and Pedodontics, Dental School, University Medicine Greifswald, Greifswald, Germany
| | - S. Offenbacher
- University of North Carolina–Chapel Hill, School of Dentistry, Department of Periodontology, Chapel Hill, NC, USA
| | - K. Divaris
- University of North Carolina–Chapel Hill, School of Dentistry, Department of Pediatric Dentistry, Chapel Hill, NC, USA
- University of North Carolina–Chapel Hill, Gillings School of Global Public Health, Department of Epidemiology, Chapel Hill, NC, USA
| | - A. Franke
- Institute of Clinical Molecular Biology, Christian-Albrechts-University, Kiel, Germany
| | - S. Schreiber
- Clinic of Internal Medicine, University Clinic Schleswig-Holstein, Kiel, Germany
- Institute of Clinical Molecular Biology, Christian-Albrechts-University, Kiel, Germany
| | - H. Dommisch
- Department of Periodontology and Synoptic Medicine, Institute for Dental and Craniofacial Sciences, Charité–University Medicine Berlin, Berlin, Germany
| | - E. Weiss
- Maurice and Gabriella Goldschleger School of Dental Medicine, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - A.S. Schaefer
- Department of Periodontology and Synoptic Medicine, Institute for Dental and Craniofacial Sciences, Charité–University Medicine Berlin, Berlin, Germany
| | - Y. Houri-Haddad
- Department of Prosthodontics, Hadassah Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel
| | - F.A. Iraqi
- Department of Clinical Microbiology and Immunology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
240
|
Stortz JA, Raymond SL, Mira JC, Moldawer LL, Mohr AM, Efron PA. Murine Models of Sepsis and Trauma: Can We Bridge the Gap? ILAR J 2017; 58:90-105. [PMID: 28444204 PMCID: PMC5886315 DOI: 10.1093/ilar/ilx007] [Citation(s) in RCA: 112] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 02/20/2017] [Accepted: 02/23/2017] [Indexed: 02/06/2023] Open
Abstract
Sepsis and trauma are both leading causes of death in the United States and represent major public health challenges. Murine models have largely been used in sepsis and trauma research to better understand the pathophysiological changes that occur after an insult and to develop potential life-saving therapeutic agents. Mice are favorable subjects for this type of research given the variety of readily available strains including inbred, outbred, and transgenic strains. In addition, they are relatively easy to maintain and have a high fecundity. However, pharmacological therapies demonstrating promise in preclinical mouse models of sepsis and trauma often fail to demonstrate similar efficacy in human clinical trials, prompting considerable criticism surrounding the capacity of murine models to recapitulate complex human diseases like sepsis and traumatic injury. Fundamental differences between the two species include, but are not limited to, the divergence of the transcriptomic response, the mismatch of temporal response patterns, differences in both innate and adaptive immunity, and heterogeneity within the human population in comparison to the homogeneity of highly inbred mouse strains. Given the ongoing controversy, this narrative review aims to not only highlight the historical importance of the mouse as an animal research model but also highlight the current benefits and limitations of the model as it pertains to sepsis and trauma. Lastly, this review will propose future directions that may promote further use of the model.
Collapse
Affiliation(s)
- Julie A. Stortz
- Julie A. Stortz, MD, is a research fellow at the University of Florida Health Shands Hospital in Gainesville, Florida. Steven L. Raymond, MD, is a research fellow at the University of Florida Health Shands Hospital in Gainesville, Florida. Juan C. Mira, MD, is a research fellow at the University of Florida Health Shands Hospital in Gainesville, Florida. Lyle L. Moldawer, PhD, is the Robert H. and Kathleen M. Axline Basic Science Professor of Surgery at the University of Florida College of Medicine in Gainesville, FL. Alicia M. Mohr, MD, is an Associate Professor of Surgery at the University of Florida College of Medicine in Gainesville, FL. Philip A. Efron, MD, is an Associate Professor of Surgery and Anesthesiology at the University of Florida College of Medicine and Medical Director for the surgical intensive care unit at the University of Florida Health Shands Hospital, Department of Surgery, University of Florida College of Medicine, Gainesville, FL.
| | - Steven L. Raymond
- Julie A. Stortz, MD, is a research fellow at the University of Florida Health Shands Hospital in Gainesville, Florida. Steven L. Raymond, MD, is a research fellow at the University of Florida Health Shands Hospital in Gainesville, Florida. Juan C. Mira, MD, is a research fellow at the University of Florida Health Shands Hospital in Gainesville, Florida. Lyle L. Moldawer, PhD, is the Robert H. and Kathleen M. Axline Basic Science Professor of Surgery at the University of Florida College of Medicine in Gainesville, FL. Alicia M. Mohr, MD, is an Associate Professor of Surgery at the University of Florida College of Medicine in Gainesville, FL. Philip A. Efron, MD, is an Associate Professor of Surgery and Anesthesiology at the University of Florida College of Medicine and Medical Director for the surgical intensive care unit at the University of Florida Health Shands Hospital, Department of Surgery, University of Florida College of Medicine, Gainesville, FL.
| | - Juan C. Mira
- Julie A. Stortz, MD, is a research fellow at the University of Florida Health Shands Hospital in Gainesville, Florida. Steven L. Raymond, MD, is a research fellow at the University of Florida Health Shands Hospital in Gainesville, Florida. Juan C. Mira, MD, is a research fellow at the University of Florida Health Shands Hospital in Gainesville, Florida. Lyle L. Moldawer, PhD, is the Robert H. and Kathleen M. Axline Basic Science Professor of Surgery at the University of Florida College of Medicine in Gainesville, FL. Alicia M. Mohr, MD, is an Associate Professor of Surgery at the University of Florida College of Medicine in Gainesville, FL. Philip A. Efron, MD, is an Associate Professor of Surgery and Anesthesiology at the University of Florida College of Medicine and Medical Director for the surgical intensive care unit at the University of Florida Health Shands Hospital, Department of Surgery, University of Florida College of Medicine, Gainesville, FL.
| | - Lyle L. Moldawer
- Julie A. Stortz, MD, is a research fellow at the University of Florida Health Shands Hospital in Gainesville, Florida. Steven L. Raymond, MD, is a research fellow at the University of Florida Health Shands Hospital in Gainesville, Florida. Juan C. Mira, MD, is a research fellow at the University of Florida Health Shands Hospital in Gainesville, Florida. Lyle L. Moldawer, PhD, is the Robert H. and Kathleen M. Axline Basic Science Professor of Surgery at the University of Florida College of Medicine in Gainesville, FL. Alicia M. Mohr, MD, is an Associate Professor of Surgery at the University of Florida College of Medicine in Gainesville, FL. Philip A. Efron, MD, is an Associate Professor of Surgery and Anesthesiology at the University of Florida College of Medicine and Medical Director for the surgical intensive care unit at the University of Florida Health Shands Hospital, Department of Surgery, University of Florida College of Medicine, Gainesville, FL.
| | - Alicia M. Mohr
- Julie A. Stortz, MD, is a research fellow at the University of Florida Health Shands Hospital in Gainesville, Florida. Steven L. Raymond, MD, is a research fellow at the University of Florida Health Shands Hospital in Gainesville, Florida. Juan C. Mira, MD, is a research fellow at the University of Florida Health Shands Hospital in Gainesville, Florida. Lyle L. Moldawer, PhD, is the Robert H. and Kathleen M. Axline Basic Science Professor of Surgery at the University of Florida College of Medicine in Gainesville, FL. Alicia M. Mohr, MD, is an Associate Professor of Surgery at the University of Florida College of Medicine in Gainesville, FL. Philip A. Efron, MD, is an Associate Professor of Surgery and Anesthesiology at the University of Florida College of Medicine and Medical Director for the surgical intensive care unit at the University of Florida Health Shands Hospital, Department of Surgery, University of Florida College of Medicine, Gainesville, FL.
| | - Philip A. Efron
- Julie A. Stortz, MD, is a research fellow at the University of Florida Health Shands Hospital in Gainesville, Florida. Steven L. Raymond, MD, is a research fellow at the University of Florida Health Shands Hospital in Gainesville, Florida. Juan C. Mira, MD, is a research fellow at the University of Florida Health Shands Hospital in Gainesville, Florida. Lyle L. Moldawer, PhD, is the Robert H. and Kathleen M. Axline Basic Science Professor of Surgery at the University of Florida College of Medicine in Gainesville, FL. Alicia M. Mohr, MD, is an Associate Professor of Surgery at the University of Florida College of Medicine in Gainesville, FL. Philip A. Efron, MD, is an Associate Professor of Surgery and Anesthesiology at the University of Florida College of Medicine and Medical Director for the surgical intensive care unit at the University of Florida Health Shands Hospital, Department of Surgery, University of Florida College of Medicine, Gainesville, FL.
| |
Collapse
|
241
|
Li B, Qing T, Zhu J, Wen Z, Yu Y, Fukumura R, Zheng Y, Gondo Y, Shi L. A Comprehensive Mouse Transcriptomic BodyMap across 17 Tissues by RNA-seq. Sci Rep 2017; 7:4200. [PMID: 28646208 PMCID: PMC5482823 DOI: 10.1038/s41598-017-04520-z] [Citation(s) in RCA: 111] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Accepted: 05/16/2017] [Indexed: 02/07/2023] Open
Abstract
The mouse has been widely used as a model organism for studying human diseases and for evaluating drug safety and efficacy. Many diseases and drug effects exhibit tissue specificity that may be reflected by tissue-specific gene-expression profiles. Here we construct a comprehensive mouse transcriptomic BodyMap across 17 tissues of six-weeks old C57BL/6JJcl mice using RNA-seq. We find different expression patterns between protein-coding and non-coding genes. Liver expressed the least complex transcriptomes, that is, the smallest number of genes detected in liver across all 17 tissues, whereas testis and ovary harbor more complex transcriptomes than other tissues. We report a comprehensive list of tissue-specific genes across 17 tissues, along with a list of 4,781 housekeeping genes in mouse. In addition, we propose a list of 27 consistently and highly expressed genes that can be used as reference controls in expression-profiling analysis. Our study provides a unique resource of mouse gene-expression profiles, which is helpful for further biomedical research.
Collapse
Affiliation(s)
- Bin Li
- Center for Pharmacogenomics, School of Pharmacy, and State Key Laboratory of Genetic Engineering, School of Life Sciences and Shanghai Cancer Hospital/Cancer Institute, Fudan University, Shanghai, 200438, China
- Collaborative Innovation Center for Genetics and Development, Fudan University, Shanghai, 200438, China
| | - Tao Qing
- Center for Pharmacogenomics, School of Pharmacy, and State Key Laboratory of Genetic Engineering, School of Life Sciences and Shanghai Cancer Hospital/Cancer Institute, Fudan University, Shanghai, 200438, China
- Collaborative Innovation Center for Genetics and Development, Fudan University, Shanghai, 200438, China
| | - Jinhang Zhu
- Center for Pharmacogenomics, School of Pharmacy, and State Key Laboratory of Genetic Engineering, School of Life Sciences and Shanghai Cancer Hospital/Cancer Institute, Fudan University, Shanghai, 200438, China
| | - Zhuo Wen
- Center for Pharmacogenomics, School of Pharmacy, and State Key Laboratory of Genetic Engineering, School of Life Sciences and Shanghai Cancer Hospital/Cancer Institute, Fudan University, Shanghai, 200438, China
- College of Chemistry, Sichuan University, Chengdu, 610064, China
| | - Ying Yu
- Center for Pharmacogenomics, School of Pharmacy, and State Key Laboratory of Genetic Engineering, School of Life Sciences and Shanghai Cancer Hospital/Cancer Institute, Fudan University, Shanghai, 200438, China
- Collaborative Innovation Center for Genetics and Development, Fudan University, Shanghai, 200438, China
| | - Ryutaro Fukumura
- Mutagenesis and Genomics Team, RIKEN BioResource Center, Tsukuba, Ibaraki, 305-0074, Japan
| | - Yuanting Zheng
- Center for Pharmacogenomics, School of Pharmacy, and State Key Laboratory of Genetic Engineering, School of Life Sciences and Shanghai Cancer Hospital/Cancer Institute, Fudan University, Shanghai, 200438, China.
- Collaborative Innovation Center for Genetics and Development, Fudan University, Shanghai, 200438, China.
| | - Yoichi Gondo
- Mutagenesis and Genomics Team, RIKEN BioResource Center, Tsukuba, Ibaraki, 305-0074, Japan.
| | - Leming Shi
- Center for Pharmacogenomics, School of Pharmacy, and State Key Laboratory of Genetic Engineering, School of Life Sciences and Shanghai Cancer Hospital/Cancer Institute, Fudan University, Shanghai, 200438, China.
- Collaborative Innovation Center for Genetics and Development, Fudan University, Shanghai, 200438, China.
| |
Collapse
|
242
|
Xiao Y, Lu W, Li X, Zhao P, Yao Y, Wang X, Wang Y, Lin Z, Yu Y, Hua S, Wang L. An oligodeoxynucleotide with AAAG repeats significantly attenuates burn-induced systemic inflammatory responses via inhibiting interferon regulatory factor 5 pathway. Mol Med 2017; 23:166-176. [PMID: 28620671 DOI: 10.2119/molmed.2016.00243] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2016] [Accepted: 06/06/2017] [Indexed: 12/16/2022] Open
Abstract
Previously, we showed that an oligodeoxynucleotide with AAAG repeats (AAAG ODN) rescued mice from fatal acute lung injury (ALI) induced by influenza virus and inhibited production of tumor necrosis factor-α (TNF-α) in the injured lungs. However, the underlying mechanisms remain to be elucidated. Upon the bioinformatic analysis revealing that the AAAG ODN is consensus to interferon regulatory factor 5 (IRF5) binding site in the cis-regulatory elements of proinflammatory cytokines, we tried to explore whether the AAAG ODN could attenuate burn injury induced systemic inflammatory responses via inhibiting IRF5 pathway. Using the mouse model with sterile systemic inflammation induced by burn injury, we found that AAAG ODN prolonged the life span of the mice, decreased the expression of IRF5 at injured skin, reduced the production of TNF-α and IL-6 in blood and injured skin, and attenuated the ALI. Furthermore, AAAG ODN could bind IRF5 and inhibit the nuclear translocation of IRF5 in THP-1 cells. The data suggested that the AAAG ODN could act as a cytoplasmic decoy capable of interfering the function of IRF5, and be developed as a drug candidate for the treatment of inflammatory diseases.
Collapse
Affiliation(s)
- Yue Xiao
- Department of Molecular Biology in College of Basic Medical Sciences and Institute of Pediatrics in First Hospital, Norman Bethune Health Science Center, Jilin University, Changchun, 130021, China
| | - Wenting Lu
- Department of Molecular Biology in College of Basic Medical Sciences and Institute of Pediatrics in First Hospital, Norman Bethune Health Science Center, Jilin University, Changchun, 130021, China
| | - Xin Li
- Department of Molecular Biology in College of Basic Medical Sciences and Institute of Pediatrics in First Hospital, Norman Bethune Health Science Center, Jilin University, Changchun, 130021, China
| | - Peiyan Zhao
- Department of Molecular Biology in College of Basic Medical Sciences and Institute of Pediatrics in First Hospital, Norman Bethune Health Science Center, Jilin University, Changchun, 130021, China
| | - Yun Yao
- Department of Molecular Biology in College of Basic Medical Sciences and Institute of Pediatrics in First Hospital, Norman Bethune Health Science Center, Jilin University, Changchun, 130021, China
| | - Xiaohong Wang
- Department of Respiratory Medicine, The First Hospital of Jilin University, Norman Bethune Health Science Center, Jilin University, Changchun, 130021, China
| | - Ying Wang
- Department of Molecular Biology in College of Basic Medical Sciences and Institute of Pediatrics in First Hospital, Norman Bethune Health Science Center, Jilin University, Changchun, 130021, China
| | - Zhipeng Lin
- Department of Molecular Biology in College of Basic Medical Sciences and Institute of Pediatrics in First Hospital, Norman Bethune Health Science Center, Jilin University, Changchun, 130021, China
| | - Yongli Yu
- Department of Immunology, College of Basic Medical Sciences, Norman Bethune Health Science Center, Jilin University, Changchun, 130021, China
| | - Shucheng Hua
- Department of Respiratory Medicine, The First Hospital of Jilin University, Norman Bethune Health Science Center, Jilin University, Changchun, 130021, China
| | - Liying Wang
- Department of Immunology, College of Basic Medical Sciences, Norman Bethune Health Science Center, Jilin University, Changchun, 130021, China
| |
Collapse
|
243
|
Schwager J, Richard N, Widmer F, Raederstorff D. Resveratrol distinctively modulates the inflammatory profiles of immune and endothelial cells. Altern Ther Health Med 2017; 17:309. [PMID: 28610607 PMCID: PMC5470273 DOI: 10.1186/s12906-017-1823-z] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 06/06/2017] [Indexed: 12/26/2022]
Abstract
Background The phenolic substance resveratrol (RES) is a plant metabolite known to modulate numerous physiological functions and to exert beneficial effects as a cancer-chemopreventing agent and on neurological, hepatic, and cardiovascular systems. Since the compound affects the lifespan of yeast and flies it might be an anti-aging substance. Mechanistically, RES is involved in down regulating the inflammatory response. The pleiotropic effects of RES in cells of the immune and endothelial system were examined in this study. Results Murine macrophages (RAW264.7 cells), human monocytic/leukemia cells (THP-1), PBLs and HUVECs were incubated with RES and activated with inflammatory stimuli such as LPS or TNF-α. Inflammatory mediators and adhesion molecules were measured by multiplex analysis and gene expression was quantified by RT-PCR. In PBLs, which were activated with LPS, RES blunted the production of TNF-α, CCL2/MCP-1, CCL5/RANTES, CXCL8/IL-8, whereas it increased the production of IL-1β, IL-6, CCL4/MIP-1β and CXCL10/IP-10. Thus, in the blood compartment chemokines attracting mainly monocytes were up-regulated by RES, while those attracting T lymphocytes or neutrophils were diminished. At conditions of endothelial dysfunction (ED), RES reduced the expression of cytokines, chemokines, ICAM and GM-CSF in TNF-α activated HUVECs, whereas eNOS expression was corrected to pre-ED homeostasis. In macrophages nitric oxide, PGE2, cytokines (TNF-α, IL-1β, IL-6) and chemokines (CCL2/MCP-1, CCL4/MIP-1β, CCL5/RANTES, CXCL10/IP-10) were reduced by the phenolic substance. Conclusions RES had cell-specific and context-dependent effects, in particular on the expression of IL-1β, IL-6, CCL4/MIP-1β and CXCL10/IP-10. It enhanced cellular features that mirror increased alertness to disturbed immune homeostasis in the vascular-endothelial compartment (e.g. increased production of IL-1β or IL-6), whereas it blunted inflammatory mediators in macrophages and consequently chronic inflammation. We infer from the present in vitro study, that RES has unique properties in the regulation of inflammatory and immune responses, which are controlled in a complex hierarchical and temporal order. Electronic supplementary material The online version of this article (doi:10.1186/s12906-017-1823-z) contains supplementary material, which is available to authorized users.
Collapse
|
244
|
Ehret T, Torelli F, Klotz C, Pedersen AB, Seeber F. Translational Rodent Models for Research on Parasitic Protozoa-A Review of Confounders and Possibilities. Front Cell Infect Microbiol 2017. [PMID: 28638807 PMCID: PMC5461347 DOI: 10.3389/fcimb.2017.00238] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Rodents, in particular Mus musculus, have a long and invaluable history as models for human diseases in biomedical research, although their translational value has been challenged in a number of cases. We provide some examples in which rodents have been suboptimal as models for human biology and discuss confounders which influence experiments and may explain some of the misleading results. Infections of rodents with protozoan parasites are no exception in requiring close consideration upon model choice. We focus on the significant differences between inbred, outbred and wild animals, and the importance of factors such as microbiota, which are gaining attention as crucial variables in infection experiments. Frequently, mouse or rat models are chosen for convenience, e.g., availability in the institution rather than on an unbiased evaluation of whether they provide the answer to a given question. Apart from a general discussion on translational success or failure, we provide examples where infections with single-celled parasites in a chosen lab rodent gave contradictory or misleading results, and when possible discuss the reason for this. We present emerging alternatives to traditional rodent models, such as humanized mice and organoid primary cell cultures. So-called recombinant inbred strains such as the Collaborative Cross collection are also a potential solution for certain challenges. In addition, we emphasize the advantages of using wild rodents for certain immunological, ecological, and/or behavioral questions. The experimental challenges (e.g., availability of species-specific reagents) that come with the use of such non-model systems are also discussed. Our intention is to foster critical judgment of both traditional and newly available translational rodent models for research on parasitic protozoa that can complement the existing mouse and rat models.
Collapse
Affiliation(s)
- Totta Ehret
- FG16 - Mycotic and Parasitic Agents and Mycobacteria, Robert Koch InstituteBerlin, Germany.,Department of Molecular Parasitology, Humboldt-Universität zu BerlinBerlin, Germany
| | - Francesca Torelli
- FG16 - Mycotic and Parasitic Agents and Mycobacteria, Robert Koch InstituteBerlin, Germany
| | - Christian Klotz
- FG16 - Mycotic and Parasitic Agents and Mycobacteria, Robert Koch InstituteBerlin, Germany
| | - Amy B Pedersen
- School of Biological Sciences, University of EdinburghEdinburgh, United Kingdom
| | - Frank Seeber
- FG16 - Mycotic and Parasitic Agents and Mycobacteria, Robert Koch InstituteBerlin, Germany
| |
Collapse
|
245
|
Steele AM, Starr ME, Saito H. Late Therapeutic Intervention with Antibiotics and Fluid Resuscitation Allows for a Prolonged Disease Course with High Survival in a Severe Murine Model of Sepsis. Shock 2017; 47:726-734. [PMID: 27879561 PMCID: PMC5432399 DOI: 10.1097/shk.0000000000000799] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Current animal models of sepsis often incorporate antibiotics to be consistent with clinical standards for treatment of patients in the intensive care unit. However, such experimental intervention is commonly initiated very early after infectious insult, which likely blunts the progression of systemic inflammation and downstream pathology. The objective of this study was to establish an animal model of sepsis with delayed therapeutic intervention, allowing a longer disease course and downstream pathology, but still resulting in a high survival rate. Severe lethal abdominal infection was initiated in young adult (17-18-week-old) C57BL/6 mice by cecal slurry (CS) injection. When initiated early (1- or 6-h post-CS injection), antibiotic treatment (imipenem, 1.5 mg/mouse i.p., twice/day for 5 days) rescued the majority of mice; however, few of these mice showed evidence of bacteremia, cytokinemia, or organ injury. When antibiotic treatment was delayed until late time-points (12- or 24-h post-CS injection) the majority of animals did not survive beyond 48 h. When fluid resuscitation (physiological saline, s.c.) was performed in combination with antibiotic treatment (twice daily) beginning at these late time-points, the majority of mice survived (75%) and showed bacteremia, cytokinemia, organ dysfunction, and prolonged body weight loss (<90% for 4 weeks). We recommend that this new repeated combination treatment with antibiotics and fluids resuscitation be initiated at a late time point after bacteremia becomes evident because this model more closely mimics the downstream pathological characteristics of severe clinical sepsis yet maintains a high survival rate. This model would be advantageous for studies on severe sepsis and postintensive care illness.
Collapse
Affiliation(s)
- Allison M. Steele
- Department of Physiology, University of Kentucky, Lexington, KY 40536
| | - Marlene E. Starr
- Department of Surgery, University of Kentucky, Lexington, KY 40536
| | - Hiroshi Saito
- Department of Physiology, University of Kentucky, Lexington, KY 40536
- Department of Surgery, University of Kentucky, Lexington, KY 40536
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536
| |
Collapse
|
246
|
Haworth KG, Ironside C, Norgaard ZK, Obenza WM, Adair JE, Kiem HP. In Vivo Murine-Matured Human CD3 + Cells as a Preclinical Model for T Cell-Based Immunotherapies. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2017. [PMID: 28649577 PMCID: PMC5470556 DOI: 10.1016/j.omtm.2017.05.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Adoptive cellular immunotherapy is a promising and powerful method for the treatment of a broad range of malignant and infectious diseases. Although the concept of cellular immunotherapy was originally proposed in the 1990s, it has not seen successful clinical application until recent years. Despite significant progress in creating engineered receptors against both malignant and viral epitopes, no efficient preclinical animal models exist for rapidly testing and directly comparing these engineered receptors. The use of matured human T cells in mice usually leads to graft-versus-host disease (GvHD), which severely limits the effectiveness of such studies. Alternatively, adult apheresis CD34+ cells engraft in neonatal non-obese diabetic (NOD)-severe combined immunodeficiency (SCID)-common γ chain–/– (NSG) mice and lead to the development of CD3+ T cells in peripheral circulation. We demonstrate that these in vivo murine-matured autologous CD3+ T cells from humans (MATCH) can be collected from the mice, engineered with lentiviral vectors, reinfused into the mice, and detected in multiple lymphoid compartments at stable levels over 50 days after injection. Unlike autologous CD3+ cells collected from human donors, these MATCH mice did not exhibit GvHD after T cell administration. This novel mouse model offers the opportunity to screen different immunotherapy-based treatments in a preclinical setting.
Collapse
Affiliation(s)
- Kevin G Haworth
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109-1024, USA
| | - Christina Ironside
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109-1024, USA
| | - Zachary K Norgaard
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109-1024, USA
| | - Willimark M Obenza
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109-1024, USA
| | - Jennifer E Adair
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109-1024, USA.,Department of Medicine, University of Washington, Seattle, WA 98195, USA
| | - Hans-Peter Kiem
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109-1024, USA.,Department of Medicine, University of Washington, Seattle, WA 98195, USA.,Department of Pathology, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
247
|
Millar LJ, Shi L, Hoerder-Suabedissen A, Molnár Z. Neonatal Hypoxia Ischaemia: Mechanisms, Models, and Therapeutic Challenges. Front Cell Neurosci 2017; 11:78. [PMID: 28533743 PMCID: PMC5420571 DOI: 10.3389/fncel.2017.00078] [Citation(s) in RCA: 231] [Impact Index Per Article: 28.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Accepted: 03/07/2017] [Indexed: 12/11/2022] Open
Abstract
Neonatal hypoxia-ischaemia (HI) is the most common cause of death and disability in human neonates, and is often associated with persistent motor, sensory, and cognitive impairment. Improved intensive care technology has increased survival without preventing neurological disorder, increasing morbidity throughout the adult population. Early preventative or neuroprotective interventions have the potential to rescue brain development in neonates, yet only one therapeutic intervention is currently licensed for use in developed countries. Recent investigations of the transient cortical layer known as subplate, especially regarding subplate's secretory role, opens up a novel set of potential molecular modulators of neonatal HI injury. This review examines the biological mechanisms of human neonatal HI, discusses evidence for the relevance of subplate-secreted molecules to this condition, and evaluates available animal models. Neuroserpin, a neuronally released neuroprotective factor, is discussed as a case study for developing new potential pharmacological interventions for use post-ischaemic injury.
Collapse
Affiliation(s)
- Lancelot J. Millar
- Molnár Group, Department of Physiology, Anatomy and Genetics, University of OxfordOxford, UK
| | - Lei Shi
- Molnár Group, Department of Physiology, Anatomy and Genetics, University of OxfordOxford, UK
- JNU-HKUST Joint Laboratory for Neuroscience and Innovative Drug Research, College of Pharmacy, Jinan UniversityGuangzhou, China
| | | | - Zoltán Molnár
- Molnár Group, Department of Physiology, Anatomy and Genetics, University of OxfordOxford, UK
| |
Collapse
|
248
|
Parker D. Humanized Mouse Models of Staphylococcus aureus Infection. Front Immunol 2017; 8:512. [PMID: 28523002 PMCID: PMC5415562 DOI: 10.3389/fimmu.2017.00512] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 04/18/2017] [Indexed: 12/18/2022] Open
Abstract
Staphylococcus aureus is a successful human pathogen that has adapted itself in response to selection pressure by the human immune system. A commensal of the human skin and nose, it is a leading cause of several conditions: skin and soft tissue infection, pneumonia, septicemia, peritonitis, bacteremia, and endocarditis. Mice have been used extensively in all these conditions to identify virulence factors and host components important for pathogenesis. Although significant effort has gone toward development of an anti-staphylococcal vaccine, antibodies have proven ineffective in preventing infection in humans after successful studies in mice. These results have raised questions as to the utility of mice to predict patient outcome and suggest that humanized mice might prove useful in modeling infection. The development of humanized mouse models of S. aureus infection will allow us to assess the contribution of several human-specific virulence factors, in addition to exploring components of the human immune system in protection against S. aureus infection. Their use is discussed in light of several recently reported studies.
Collapse
Affiliation(s)
- Dane Parker
- Department of Pediatrics, Columbia University, New York, NY, USA
| |
Collapse
|
249
|
Cuenda A, Sanz-Ezquerro JJ. p38γ and p38δ: From Spectators to Key Physiological Players. Trends Biochem Sci 2017; 42:431-442. [PMID: 28473179 DOI: 10.1016/j.tibs.2017.02.008] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 02/14/2017] [Accepted: 02/22/2017] [Indexed: 12/20/2022]
Abstract
Although the physiological roles of p38γ and p38δ signalling pathways are largely unknown, new genetic and pharmacological tools are providing groundbreaking information on the function of these two stress-activated protein kinases. Recent studies show the importance of p38γ and p38δ in the regulation of processes as diverse as cytokine production, protein synthesis, exocytosis, cell migration, gene expression, and neuron activity, which have an acute impact on the development of pathologies related to inflammation, diabetes, neurodegeneration, and cancer. These recent breakthroughs are resolving some of the questions that have long been asked regarding the function of p38γ and p38δ in biology and pathology.
Collapse
Affiliation(s)
- Ana Cuenda
- Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC, Campus de Cantoblanco, 28049 Madrid, Spain.
| | - Juan José Sanz-Ezquerro
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología/CSIC, Campus de Cantoblanco, 28049 Madrid, Spain
| |
Collapse
|
250
|
Do we need animal models to advance research on inflammatory joint disease? Joint Bone Spine 2017; 84:381-383. [PMID: 28456601 DOI: 10.1016/j.jbspin.2017.04.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/09/2017] [Indexed: 11/20/2022]
|