301
|
Skrypnyk NI, Siskind LJ, Faubel S, de Caestecker MP. Bridging translation for acute kidney injury with better preclinical modeling of human disease. Am J Physiol Renal Physiol 2016; 310:F972-84. [PMID: 26962107 PMCID: PMC4889323 DOI: 10.1152/ajprenal.00552.2015] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Accepted: 03/04/2016] [Indexed: 12/14/2022] Open
Abstract
The current lack of effective therapeutics for patients with acute kidney injury (AKI) represents an important and unmet medical need. Given the importance of the clinical problem, it is time for us to take a few steps back and reexamine current practices. The focus of this review is to explore the extent to which failure of therapeutic translation from animal studies to human studies stems from deficiencies in the preclinical models of AKI. We will evaluate whether the preclinical models of AKI that are commonly used recapitulate the known pathophysiologies of AKI that are being modeled in humans, focusing on four common scenarios that are studied in clinical therapeutic intervention trials: cardiac surgery-induced AKI; contrast-induced AKI; cisplatin-induced AKI; and sepsis associated AKI. Based on our observations, we have identified a number of common limitations in current preclinical modeling of AKI that could be addressed. In the long term, we suggest that progress in developing better preclinical models of AKI will depend on developing a better understanding of human AKI. To this this end, we suggest that there is a need to develop greater in-depth molecular analyses of kidney biopsy tissues coupled with improved clinical and molecular classification of patients with AKI.
Collapse
Affiliation(s)
- Nataliya I Skrypnyk
- Division of Nephology and Hypertension, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Leah J Siskind
- Department of Pharmacology and Toxicology, James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky; and
| | - Sarah Faubel
- Renal Division, University of Colorado Denver and Denver Veterans Affairs Medical Center, Aurora, Colorado
| | - Mark P de Caestecker
- Division of Nephology and Hypertension, Vanderbilt University Medical Center, Nashville, Tennessee;
| |
Collapse
|
302
|
De Filippis L, Halikere A, McGowan H, Moore JC, Tischfield JA, Hart RP, Pang ZP. Ethanol-mediated activation of the NLRP3 inflammasome in iPS cells and iPS cells-derived neural progenitor cells. Mol Brain 2016; 9:51. [PMID: 27160314 PMCID: PMC4862119 DOI: 10.1186/s13041-016-0221-7] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Accepted: 04/08/2016] [Indexed: 12/19/2022] Open
Abstract
Background Alcohol abuse produces an enormous impact on health, society, and the economy. Currently, there are very limited therapies available, largely due to the poor understanding of mechanisms underlying alcohol use disorders (AUDs) in humans. Oxidative damage of mitochondria and cellular proteins aggravates the progression of neuroinflammation and neurological disorders initiated by alcohol abuse. Results Here we show that ethanol exposure causes neuroinflammation in both human induced pluripotent stem (iPS) cells and human neural progenitor cells (NPCs). Ethanol exposure for 24 hours or 7 days does not affect the proliferation of iPS cells and NPCs, but primes an innate immune-like response by activating the NLR family pyrin domain containing 3 (NLRP3) inflammasome pathway. This leads to an increase of microtubule-associated protein 1A/1B-light chain 3+ (LC3B+) autophagic puncta and impairment of the mitochondrial and lysosomal distribution. In addition, a decrease of mature neurons derived from differentiating NPCs is evident in ethanol pre-exposed compared to control NPCs. Moreover, a second insult of a pro-inflammatory factor in addition to ethanol preexposure enhances innate cellular inflammation in human iPS cells. Conclusions This study provides strong evidence that neuronal inflammation contributes to the pathophysiology of AUDs through the activation of the inflammasome pathway in human cellular models.
Collapse
Affiliation(s)
- Lidia De Filippis
- Child Health Institute of New Jersey, Rutgers University-Robert Wood Johnson Medical School, room 3233D, 89 French Street, New Brunswick, NJ, 08901, USA. .,Department of Neuroscience and Cell Biology, Rutgers University-Robert Wood Johnson Medical School, room 3233D, 89 French Street, New Brunswick, NJ, 08901, USA.
| | - Apoorva Halikere
- Child Health Institute of New Jersey, Rutgers University-Robert Wood Johnson Medical School, room 3233D, 89 French Street, New Brunswick, NJ, 08901, USA.,Department of Neuroscience and Cell Biology, Rutgers University-Robert Wood Johnson Medical School, room 3233D, 89 French Street, New Brunswick, NJ, 08901, USA
| | - Heather McGowan
- Child Health Institute of New Jersey, Rutgers University-Robert Wood Johnson Medical School, room 3233D, 89 French Street, New Brunswick, NJ, 08901, USA.,Department of Neuroscience and Cell Biology, Rutgers University-Robert Wood Johnson Medical School, room 3233D, 89 French Street, New Brunswick, NJ, 08901, USA
| | - Jennifer C Moore
- Department of Genetics, Rutgers University, Piscataway, 08854, USA.,Human Genetic Institute of New Jersey, Rutgers University, Piscataway, 08854, USA
| | - Jay A Tischfield
- Department of Genetics, Rutgers University, Piscataway, 08854, USA.,Human Genetic Institute of New Jersey, Rutgers University, Piscataway, 08854, USA
| | - Ronald P Hart
- Human Genetic Institute of New Jersey, Rutgers University, Piscataway, 08854, USA.,Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, 08854, USA
| | - Zhiping P Pang
- Child Health Institute of New Jersey, Rutgers University-Robert Wood Johnson Medical School, room 3233D, 89 French Street, New Brunswick, NJ, 08901, USA. .,Department of Neuroscience and Cell Biology, Rutgers University-Robert Wood Johnson Medical School, room 3233D, 89 French Street, New Brunswick, NJ, 08901, USA.
| |
Collapse
|
303
|
Mouse Sperm Cryopreservation and Recovery of Genetically Modified Mice. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2016; 1438:55-66. [PMID: 27150083 DOI: 10.1007/978-1-4939-3661-8_3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Highly definable genetically, the humble mouse is the "reagent" mammal of choice with which to probe and begin to understand the human condition in all its complexities. With the recent advance in direct genome editing via targeted nucleases, e.g., TALEN and CRISPR/Cas9, the possibilities in using these sophisticated tools have increased substantially leading to a massive increase in the variety of strain numbers of genetically modified lines. With this increase comes a greater need to economically and creatively manage their numbers. Further, once characterized, lines may be of limited use but still need to be archived in a format allowing their rapid resurrection. Further, maintaining colonies on "the shelf" is financially draining and carries potential risks including natural disaster loss, disease, and strain contamination. Here we outline a simple and economic protocol to cryopreserve mouse sperm from many different genetic backgrounds, and outline its recovery via in vitro fertilization (IVF). The combined use of sperm cryopreservation and IVF now allows a freedom and versatility in mouse management facilitating rapid line close down with the option to later recover and rapidly expand as needed.
Collapse
|
304
|
Gonzales NM, Howell VM, Smith CM. 29th International Mammalian Genome Conference meeting report. Mamm Genome 2016; 27:169-78. [PMID: 27137837 PMCID: PMC7087999 DOI: 10.1007/s00335-016-9640-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 04/19/2016] [Indexed: 11/18/2022]
Affiliation(s)
- Natalia M Gonzales
- Department of Human Genetics, University of Chicago, Chicago, IL, 60637, USA
| | - Viive M Howell
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, Northern Sydney Local Health District, St. Leonards, NSW, 2065, Australia.,Sydney Medical School-Northern, University of Sydney, Sydney, NSW, 2006, Australia
| | - Clare M Smith
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA, 01605, USA.
| |
Collapse
|
305
|
Motwani MP, Flint JD, De Maeyer RP, Fullerton JN, Smith AM, Marks DJ, Gilroy DW. Novel translational model of resolving inflammation triggered by UV-killed E. coli. JOURNAL OF PATHOLOGY CLINICAL RESEARCH 2016; 2:154-65. [PMID: 27499924 PMCID: PMC4958736 DOI: 10.1002/cjp2.43] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Accepted: 03/05/2016] [Indexed: 11/23/2022]
Abstract
Whilst numerous studies investigating the aetiology of inflammatory diseases have been performed in rodents, the applicability of these data to human pathophysiology is frequently debated. Regardless of the strengths and weaknesses of rodent models in biomedical research, there is a need to develop models of experimental inflammation in humans. Here, we describe a self‐resolving acute inflammatory response triggered by the intradermal injection of UV‐killed Escherichia coli into the forearm of healthy volunteers. Cells and exudates were harvested from onset to resolution by applying negative pressure over the inflamed site. Onset was characterized by high blood flow, neutrophilia and peak levels of pro‐inflammatory cytokines, whilst resolution showed a decline in blood blow, reduction in neutrophils, increase in monocytes/macrophages and waning of classic pro‐inflammatory cytokine levels. An anti‐inflammatory effect, defined as suppression of onset phase events, was demonstrated by administering naproxen, a conventional non‐steroidal anti‐inflammatory drug. In summary, this model of resolving acute inflammation is minimally invasive, highly tractable and allows simultaneous investigation of the vascular response, cellular trafficking and chemical mediator profile of onset and resolution phases of acute inflammation in humans. It can serve as a translational platform to provide mechanistic insights and to test the clinical efficacy of novel anti‐inflammatory and pro‐resolving drugs, and also as a tool in patients to explore inherent defects in resolution pathways.
Collapse
Affiliation(s)
- Madhur P Motwani
- Centre for Clinical Pharmacology and Therapeutics, Division of Medicine, 5 University Street University College London London WC1E 6JF UK
| | - Julia D Flint
- Centre for Clinical Pharmacology and Therapeutics, Division of Medicine, 5 University Street University College London London WC1E 6JF UK
| | - Roel Ph De Maeyer
- Centre for Clinical Pharmacology and Therapeutics, Division of Medicine, 5 University Street University College London London WC1E 6JF UK
| | - James N Fullerton
- Centre for Clinical Pharmacology and Therapeutics, Division of Medicine, 5 University Street University College London London WC1E 6JF UK
| | - Andrew M Smith
- Microbial Diseases, Eastman Dental Institute, University College London London WC1X 8LD UK
| | - Daniel Jb Marks
- Centre for Molecular Medicine, Division of Medicine, 5 University Street University College London London WC1E 6JF UK
| | - Derek W Gilroy
- Centre for Clinical Pharmacology and Therapeutics, Division of Medicine, 5 University Street University College London London WC1E 6JF UK
| |
Collapse
|
306
|
Beura LK, Hamilton SE, Bi K, Schenkel JM, Odumade OA, Casey KA, Thompson EA, Fraser KA, Rosato PC, Filali-Mouhim A, Sekaly RP, Jenkins MK, Vezys V, Haining WN, Jameson SC, Masopust D. Normalizing the environment recapitulates adult human immune traits in laboratory mice. Nature 2016; 532:512-6. [PMID: 27096360 PMCID: PMC4871315 DOI: 10.1038/nature17655] [Citation(s) in RCA: 812] [Impact Index Per Article: 90.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 03/11/2016] [Indexed: 12/12/2022]
Abstract
Our current understanding of immunology was largely defined in laboratory mice, partly because they are inbred and genetically homogeneous, can be genetically manipulated, allow kinetic tissue analyses to be carried out from the onset of disease, and permit the use of tractable disease models. Comparably reductionist experiments are neither technically nor ethically possible in humans. However, there is growing concern that laboratory mice do not reflect relevant aspects of the human immune system, which may account for failures to translate disease treatments from bench to bedside. Laboratory mice live in abnormally hygienic specific pathogen free (SPF) barrier facilities. Here we show that standard laboratory mouse husbandry has profound effects on the immune system and that environmental changes produce mice with immune systems closer to those of adult humans. Laboratory mice--like newborn, but not adult, humans--lack effector-differentiated and mucosally distributed memory T cells. These cell populations were present in free-living barn populations of feral mice and pet store mice with diverse microbial experience, and were induced in laboratory mice after co-housing with pet store mice, suggesting that the environment is involved in the induction of these cells. Altering the living conditions of mice profoundly affected the cellular composition of the innate and adaptive immune systems, resulted in global changes in blood cell gene expression to patterns that more closely reflected the immune signatures of adult humans rather than neonates, altered resistance to infection, and influenced T-cell differentiation in response to a de novo viral infection. These data highlight the effects of environment on the basal immune state and response to infection and suggest that restoring physiological microbial exposure in laboratory mice could provide a relevant tool for modelling immunological events in free-living organisms, including humans.
Collapse
Affiliation(s)
- Lalit K Beura
- Center for Immunology, Department of Microbiology and Immunology, University of Minnesota, Minneapolis, Minnesota 55414, USA
| | - Sara E Hamilton
- Center for Immunology, Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota 55414, USA
| | - Kevin Bi
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, and Pediatric Hematology and Oncology, Children's Hospital, Boston, Massachusetts 02115, USA
| | - Jason M Schenkel
- Center for Immunology, Department of Microbiology and Immunology, University of Minnesota, Minneapolis, Minnesota 55414, USA
| | - Oludare A Odumade
- Center for Immunology, Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota 55414, USA
| | - Kerry A Casey
- Center for Immunology, Department of Microbiology and Immunology, University of Minnesota, Minneapolis, Minnesota 55414, USA
| | - Emily A Thompson
- Center for Immunology, Department of Microbiology and Immunology, University of Minnesota, Minneapolis, Minnesota 55414, USA
| | - Kathryn A Fraser
- Center for Immunology, Department of Microbiology and Immunology, University of Minnesota, Minneapolis, Minnesota 55414, USA
| | - Pamela C Rosato
- Center for Immunology, Department of Microbiology and Immunology, University of Minnesota, Minneapolis, Minnesota 55414, USA
| | - Ali Filali-Mouhim
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio 44106, USA
| | - Rafick P Sekaly
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio 44106, USA
| | - Marc K Jenkins
- Center for Immunology, Department of Microbiology and Immunology, University of Minnesota, Minneapolis, Minnesota 55414, USA
| | - Vaiva Vezys
- Center for Immunology, Department of Microbiology and Immunology, University of Minnesota, Minneapolis, Minnesota 55414, USA
| | - W Nicholas Haining
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, and Pediatric Hematology and Oncology, Children's Hospital, Boston, Massachusetts 02115, USA
| | - Stephen C Jameson
- Center for Immunology, Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota 55414, USA
| | - David Masopust
- Center for Immunology, Department of Microbiology and Immunology, University of Minnesota, Minneapolis, Minnesota 55414, USA
| |
Collapse
|
307
|
Reese TA, Bi K, Kambal A, Filali-Mouhim A, Beura LK, Bürger MC, Pulendran B, Sekaly RP, Jameson SC, Masopust D, Haining WN, Virgin HW. Sequential Infection with Common Pathogens Promotes Human-like Immune Gene Expression and Altered Vaccine Response. Cell Host Microbe 2016; 19:713-9. [PMID: 27107939 DOI: 10.1016/j.chom.2016.04.003] [Citation(s) in RCA: 187] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Revised: 03/22/2016] [Accepted: 04/01/2016] [Indexed: 12/16/2022]
Abstract
Immune responses differ between laboratory mice and humans. Chronic infection with viruses and parasites are common in humans, but are absent in laboratory mice, and thus represent potential contributors to inter-species differences in immunity. To test this, we sequentially infected laboratory mice with herpesviruses, influenza, and an intestinal helminth and compared their blood immune signatures to mock-infected mice before and after vaccination against yellow fever virus (YFV-17D). Sequential infection altered pre- and post-vaccination gene expression, cytokines, and antibodies in blood. Sequential pathogen exposure induced gene signatures that recapitulated those seen in blood from pet store-raised versus laboratory mice, and adult versus cord blood in humans. Therefore, basal and vaccine-induced murine immune responses are altered by infection with agents common outside of barrier facilities. This raises the possibility that we can improve mouse models of vaccination and immunity by selective microbial exposure of laboratory animals to mimic that of humans.
Collapse
Affiliation(s)
- Tiffany A Reese
- Departments of Immunology and Microbiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| | - Kevin Bi
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, and Pediatric Hematology and Oncology, Children's Hospital, Boston, MA 02115, and the Broad Institute of Harvard and the Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Amal Kambal
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Ali Filali-Mouhim
- Department of Pathology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Lalit K Beura
- Department of Microbiology and Immunology, Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Matheus C Bürger
- Department of Clinical Analyses and Toxicology, School of Pharmaceutical Science at University of São Paulo, São Paulo 05508, Brazil
| | - Bali Pulendran
- Emory Vaccine Center, Yerkes National Primate Research Center and Department of Pathology, Emory University, Atlanta, GA 30329, USA
| | - Rafick-Pierre Sekaly
- Department of Pathology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Stephen C Jameson
- Department of Laboratory Medicine and Pathology, Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA
| | - David Masopust
- Department of Microbiology and Immunology, Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA
| | - W Nicholas Haining
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, and Pediatric Hematology and Oncology, Children's Hospital, Boston, MA 02115, and the Broad Institute of Harvard and the Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Herbert W Virgin
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
308
|
Pasterkamp G, van der Laan SW, Haitjema S, Foroughi Asl H, Siemelink MA, Bezemer T, van Setten J, Dichgans M, Malik R, Worrall BB, Schunkert H, Samani NJ, de Kleijn DPV, Markus HS, Hoefer IE, Michoel T, de Jager SCA, Björkegren JLM, den Ruijter HM, Asselbergs FW. Human Validation of Genes Associated With a Murine Atherosclerotic Phenotype. Arterioscler Thromb Vasc Biol 2016; 36:1240-6. [PMID: 27079880 DOI: 10.1161/atvbaha.115.306958] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Accepted: 02/17/2016] [Indexed: 11/16/2022]
Abstract
OBJECTIVE The genetically modified mouse is the most commonly used animal model for studying the pathogenesis of atherosclerotic disease. We aimed to assess if mice atherosclerosis-related genes could be validated in human disease through examination of results from genome-wide association studies. APPROACH AND RESULTS We performed a systematic review to identify atherosclerosis-causing genes in mice and carried out gene-based association tests of their human orthologs for an association with human coronary artery disease and human large artery ischemic stroke. Moreover, we investigated the association of these genes with human atherosclerotic plaque characteristics. In addition, we assessed the presence of tissue-specific cis-acting expression quantitative trait loci for these genes in humans. Finally, using pathway analyses we show that the putative atherosclerosis-causing genes revealed few associations with human coronary artery disease, large artery ischemic stroke, or atherosclerotic plaque characteristics, despite the fact that the majority of these genes have cis-acting expression quantitative trait loci. CONCLUSIONS A role for genes that has been observed in mice for atherosclerotic lesion development could scarcely be confirmed by studying associations of disease development with common human genetic variants. The value of murine atherosclerotic models for selection of therapeutic targets in human disease remains unclear.
Collapse
Affiliation(s)
- Gerard Pasterkamp
- From the Laboratory of Experimental Cardiology, Division Heart and Lungs (G.P., S.W.v.d.L., S.H., M.A.S., T.B., J.v.S., I.E.H., S.C.A.d.J., H.M.d.R.), Laboratory of Clinical Chemistry and Hematology, Division Laboratories and Pharmacy (G.P.), and Division Heart and Lungs, Department of Cardiology (F.W.A.), University Medical Center Utrecht, Utrecht, The Netherlands; Vascular Biology Unit, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden (H.F.A., J.L.M.B.); Institut für Schlaganfall- und Demenzforschung (ISD) Klinikum der Universität München, Munich, Germany (M.D., R.M.); Munich Cluster for Systems Neurology (SyNergy), Munich, Germany (M.D.); Department of Neurology, University of Virginia, Charlottesville (B.B.W.); Klinik für Herz- und Kreislauferkrankungen, Deutsches Herzzentrum München, Technische Universität München, Munich, Germany (H.S.); Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK) e.V., Partner Site Munich Heart Alliance, Munich, Germany (H.S.); Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom (N.J.S.); Leicester National Institute of Health Research Biomedical Research Unit in Cardiovascular Disease, Leicester, United Kingdom (N.J.S.); Department of Surgery, National University of Singapore, Singapore (D.P.V.d.K.); Cardiovascular Research Institute (CVRI), National University Health System, Singapore, Singapore (D.P.V.d.K.); Interuniversity Cardiology Institute of the Netherlands, Utrecht, The Netherlands (D.P.V.d.K.); Department of Neurosciences, University of Cambridge, Cambridge, United Kingdom (H.S.M.); The Roslin Institute, University of Edinburgh, Edinburgh, United Kingdom (T.M.); Department of Genetics and Genomic Sciences, Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY (J.L.M.B.); Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estoni
| | - Sander W van der Laan
- From the Laboratory of Experimental Cardiology, Division Heart and Lungs (G.P., S.W.v.d.L., S.H., M.A.S., T.B., J.v.S., I.E.H., S.C.A.d.J., H.M.d.R.), Laboratory of Clinical Chemistry and Hematology, Division Laboratories and Pharmacy (G.P.), and Division Heart and Lungs, Department of Cardiology (F.W.A.), University Medical Center Utrecht, Utrecht, The Netherlands; Vascular Biology Unit, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden (H.F.A., J.L.M.B.); Institut für Schlaganfall- und Demenzforschung (ISD) Klinikum der Universität München, Munich, Germany (M.D., R.M.); Munich Cluster for Systems Neurology (SyNergy), Munich, Germany (M.D.); Department of Neurology, University of Virginia, Charlottesville (B.B.W.); Klinik für Herz- und Kreislauferkrankungen, Deutsches Herzzentrum München, Technische Universität München, Munich, Germany (H.S.); Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK) e.V., Partner Site Munich Heart Alliance, Munich, Germany (H.S.); Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom (N.J.S.); Leicester National Institute of Health Research Biomedical Research Unit in Cardiovascular Disease, Leicester, United Kingdom (N.J.S.); Department of Surgery, National University of Singapore, Singapore (D.P.V.d.K.); Cardiovascular Research Institute (CVRI), National University Health System, Singapore, Singapore (D.P.V.d.K.); Interuniversity Cardiology Institute of the Netherlands, Utrecht, The Netherlands (D.P.V.d.K.); Department of Neurosciences, University of Cambridge, Cambridge, United Kingdom (H.S.M.); The Roslin Institute, University of Edinburgh, Edinburgh, United Kingdom (T.M.); Department of Genetics and Genomic Sciences, Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY (J.L.M.B.); Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estoni
| | - Saskia Haitjema
- From the Laboratory of Experimental Cardiology, Division Heart and Lungs (G.P., S.W.v.d.L., S.H., M.A.S., T.B., J.v.S., I.E.H., S.C.A.d.J., H.M.d.R.), Laboratory of Clinical Chemistry and Hematology, Division Laboratories and Pharmacy (G.P.), and Division Heart and Lungs, Department of Cardiology (F.W.A.), University Medical Center Utrecht, Utrecht, The Netherlands; Vascular Biology Unit, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden (H.F.A., J.L.M.B.); Institut für Schlaganfall- und Demenzforschung (ISD) Klinikum der Universität München, Munich, Germany (M.D., R.M.); Munich Cluster for Systems Neurology (SyNergy), Munich, Germany (M.D.); Department of Neurology, University of Virginia, Charlottesville (B.B.W.); Klinik für Herz- und Kreislauferkrankungen, Deutsches Herzzentrum München, Technische Universität München, Munich, Germany (H.S.); Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK) e.V., Partner Site Munich Heart Alliance, Munich, Germany (H.S.); Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom (N.J.S.); Leicester National Institute of Health Research Biomedical Research Unit in Cardiovascular Disease, Leicester, United Kingdom (N.J.S.); Department of Surgery, National University of Singapore, Singapore (D.P.V.d.K.); Cardiovascular Research Institute (CVRI), National University Health System, Singapore, Singapore (D.P.V.d.K.); Interuniversity Cardiology Institute of the Netherlands, Utrecht, The Netherlands (D.P.V.d.K.); Department of Neurosciences, University of Cambridge, Cambridge, United Kingdom (H.S.M.); The Roslin Institute, University of Edinburgh, Edinburgh, United Kingdom (T.M.); Department of Genetics and Genomic Sciences, Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY (J.L.M.B.); Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estoni
| | - Hassan Foroughi Asl
- From the Laboratory of Experimental Cardiology, Division Heart and Lungs (G.P., S.W.v.d.L., S.H., M.A.S., T.B., J.v.S., I.E.H., S.C.A.d.J., H.M.d.R.), Laboratory of Clinical Chemistry and Hematology, Division Laboratories and Pharmacy (G.P.), and Division Heart and Lungs, Department of Cardiology (F.W.A.), University Medical Center Utrecht, Utrecht, The Netherlands; Vascular Biology Unit, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden (H.F.A., J.L.M.B.); Institut für Schlaganfall- und Demenzforschung (ISD) Klinikum der Universität München, Munich, Germany (M.D., R.M.); Munich Cluster for Systems Neurology (SyNergy), Munich, Germany (M.D.); Department of Neurology, University of Virginia, Charlottesville (B.B.W.); Klinik für Herz- und Kreislauferkrankungen, Deutsches Herzzentrum München, Technische Universität München, Munich, Germany (H.S.); Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK) e.V., Partner Site Munich Heart Alliance, Munich, Germany (H.S.); Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom (N.J.S.); Leicester National Institute of Health Research Biomedical Research Unit in Cardiovascular Disease, Leicester, United Kingdom (N.J.S.); Department of Surgery, National University of Singapore, Singapore (D.P.V.d.K.); Cardiovascular Research Institute (CVRI), National University Health System, Singapore, Singapore (D.P.V.d.K.); Interuniversity Cardiology Institute of the Netherlands, Utrecht, The Netherlands (D.P.V.d.K.); Department of Neurosciences, University of Cambridge, Cambridge, United Kingdom (H.S.M.); The Roslin Institute, University of Edinburgh, Edinburgh, United Kingdom (T.M.); Department of Genetics and Genomic Sciences, Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY (J.L.M.B.); Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estoni
| | - Marten A Siemelink
- From the Laboratory of Experimental Cardiology, Division Heart and Lungs (G.P., S.W.v.d.L., S.H., M.A.S., T.B., J.v.S., I.E.H., S.C.A.d.J., H.M.d.R.), Laboratory of Clinical Chemistry and Hematology, Division Laboratories and Pharmacy (G.P.), and Division Heart and Lungs, Department of Cardiology (F.W.A.), University Medical Center Utrecht, Utrecht, The Netherlands; Vascular Biology Unit, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden (H.F.A., J.L.M.B.); Institut für Schlaganfall- und Demenzforschung (ISD) Klinikum der Universität München, Munich, Germany (M.D., R.M.); Munich Cluster for Systems Neurology (SyNergy), Munich, Germany (M.D.); Department of Neurology, University of Virginia, Charlottesville (B.B.W.); Klinik für Herz- und Kreislauferkrankungen, Deutsches Herzzentrum München, Technische Universität München, Munich, Germany (H.S.); Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK) e.V., Partner Site Munich Heart Alliance, Munich, Germany (H.S.); Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom (N.J.S.); Leicester National Institute of Health Research Biomedical Research Unit in Cardiovascular Disease, Leicester, United Kingdom (N.J.S.); Department of Surgery, National University of Singapore, Singapore (D.P.V.d.K.); Cardiovascular Research Institute (CVRI), National University Health System, Singapore, Singapore (D.P.V.d.K.); Interuniversity Cardiology Institute of the Netherlands, Utrecht, The Netherlands (D.P.V.d.K.); Department of Neurosciences, University of Cambridge, Cambridge, United Kingdom (H.S.M.); The Roslin Institute, University of Edinburgh, Edinburgh, United Kingdom (T.M.); Department of Genetics and Genomic Sciences, Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY (J.L.M.B.); Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estoni
| | - Tim Bezemer
- From the Laboratory of Experimental Cardiology, Division Heart and Lungs (G.P., S.W.v.d.L., S.H., M.A.S., T.B., J.v.S., I.E.H., S.C.A.d.J., H.M.d.R.), Laboratory of Clinical Chemistry and Hematology, Division Laboratories and Pharmacy (G.P.), and Division Heart and Lungs, Department of Cardiology (F.W.A.), University Medical Center Utrecht, Utrecht, The Netherlands; Vascular Biology Unit, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden (H.F.A., J.L.M.B.); Institut für Schlaganfall- und Demenzforschung (ISD) Klinikum der Universität München, Munich, Germany (M.D., R.M.); Munich Cluster for Systems Neurology (SyNergy), Munich, Germany (M.D.); Department of Neurology, University of Virginia, Charlottesville (B.B.W.); Klinik für Herz- und Kreislauferkrankungen, Deutsches Herzzentrum München, Technische Universität München, Munich, Germany (H.S.); Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK) e.V., Partner Site Munich Heart Alliance, Munich, Germany (H.S.); Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom (N.J.S.); Leicester National Institute of Health Research Biomedical Research Unit in Cardiovascular Disease, Leicester, United Kingdom (N.J.S.); Department of Surgery, National University of Singapore, Singapore (D.P.V.d.K.); Cardiovascular Research Institute (CVRI), National University Health System, Singapore, Singapore (D.P.V.d.K.); Interuniversity Cardiology Institute of the Netherlands, Utrecht, The Netherlands (D.P.V.d.K.); Department of Neurosciences, University of Cambridge, Cambridge, United Kingdom (H.S.M.); The Roslin Institute, University of Edinburgh, Edinburgh, United Kingdom (T.M.); Department of Genetics and Genomic Sciences, Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY (J.L.M.B.); Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estoni
| | - Jessica van Setten
- From the Laboratory of Experimental Cardiology, Division Heart and Lungs (G.P., S.W.v.d.L., S.H., M.A.S., T.B., J.v.S., I.E.H., S.C.A.d.J., H.M.d.R.), Laboratory of Clinical Chemistry and Hematology, Division Laboratories and Pharmacy (G.P.), and Division Heart and Lungs, Department of Cardiology (F.W.A.), University Medical Center Utrecht, Utrecht, The Netherlands; Vascular Biology Unit, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden (H.F.A., J.L.M.B.); Institut für Schlaganfall- und Demenzforschung (ISD) Klinikum der Universität München, Munich, Germany (M.D., R.M.); Munich Cluster for Systems Neurology (SyNergy), Munich, Germany (M.D.); Department of Neurology, University of Virginia, Charlottesville (B.B.W.); Klinik für Herz- und Kreislauferkrankungen, Deutsches Herzzentrum München, Technische Universität München, Munich, Germany (H.S.); Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK) e.V., Partner Site Munich Heart Alliance, Munich, Germany (H.S.); Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom (N.J.S.); Leicester National Institute of Health Research Biomedical Research Unit in Cardiovascular Disease, Leicester, United Kingdom (N.J.S.); Department of Surgery, National University of Singapore, Singapore (D.P.V.d.K.); Cardiovascular Research Institute (CVRI), National University Health System, Singapore, Singapore (D.P.V.d.K.); Interuniversity Cardiology Institute of the Netherlands, Utrecht, The Netherlands (D.P.V.d.K.); Department of Neurosciences, University of Cambridge, Cambridge, United Kingdom (H.S.M.); The Roslin Institute, University of Edinburgh, Edinburgh, United Kingdom (T.M.); Department of Genetics and Genomic Sciences, Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY (J.L.M.B.); Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estoni
| | - Martin Dichgans
- From the Laboratory of Experimental Cardiology, Division Heart and Lungs (G.P., S.W.v.d.L., S.H., M.A.S., T.B., J.v.S., I.E.H., S.C.A.d.J., H.M.d.R.), Laboratory of Clinical Chemistry and Hematology, Division Laboratories and Pharmacy (G.P.), and Division Heart and Lungs, Department of Cardiology (F.W.A.), University Medical Center Utrecht, Utrecht, The Netherlands; Vascular Biology Unit, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden (H.F.A., J.L.M.B.); Institut für Schlaganfall- und Demenzforschung (ISD) Klinikum der Universität München, Munich, Germany (M.D., R.M.); Munich Cluster for Systems Neurology (SyNergy), Munich, Germany (M.D.); Department of Neurology, University of Virginia, Charlottesville (B.B.W.); Klinik für Herz- und Kreislauferkrankungen, Deutsches Herzzentrum München, Technische Universität München, Munich, Germany (H.S.); Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK) e.V., Partner Site Munich Heart Alliance, Munich, Germany (H.S.); Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom (N.J.S.); Leicester National Institute of Health Research Biomedical Research Unit in Cardiovascular Disease, Leicester, United Kingdom (N.J.S.); Department of Surgery, National University of Singapore, Singapore (D.P.V.d.K.); Cardiovascular Research Institute (CVRI), National University Health System, Singapore, Singapore (D.P.V.d.K.); Interuniversity Cardiology Institute of the Netherlands, Utrecht, The Netherlands (D.P.V.d.K.); Department of Neurosciences, University of Cambridge, Cambridge, United Kingdom (H.S.M.); The Roslin Institute, University of Edinburgh, Edinburgh, United Kingdom (T.M.); Department of Genetics and Genomic Sciences, Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY (J.L.M.B.); Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estoni
| | - Rainer Malik
- From the Laboratory of Experimental Cardiology, Division Heart and Lungs (G.P., S.W.v.d.L., S.H., M.A.S., T.B., J.v.S., I.E.H., S.C.A.d.J., H.M.d.R.), Laboratory of Clinical Chemistry and Hematology, Division Laboratories and Pharmacy (G.P.), and Division Heart and Lungs, Department of Cardiology (F.W.A.), University Medical Center Utrecht, Utrecht, The Netherlands; Vascular Biology Unit, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden (H.F.A., J.L.M.B.); Institut für Schlaganfall- und Demenzforschung (ISD) Klinikum der Universität München, Munich, Germany (M.D., R.M.); Munich Cluster for Systems Neurology (SyNergy), Munich, Germany (M.D.); Department of Neurology, University of Virginia, Charlottesville (B.B.W.); Klinik für Herz- und Kreislauferkrankungen, Deutsches Herzzentrum München, Technische Universität München, Munich, Germany (H.S.); Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK) e.V., Partner Site Munich Heart Alliance, Munich, Germany (H.S.); Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom (N.J.S.); Leicester National Institute of Health Research Biomedical Research Unit in Cardiovascular Disease, Leicester, United Kingdom (N.J.S.); Department of Surgery, National University of Singapore, Singapore (D.P.V.d.K.); Cardiovascular Research Institute (CVRI), National University Health System, Singapore, Singapore (D.P.V.d.K.); Interuniversity Cardiology Institute of the Netherlands, Utrecht, The Netherlands (D.P.V.d.K.); Department of Neurosciences, University of Cambridge, Cambridge, United Kingdom (H.S.M.); The Roslin Institute, University of Edinburgh, Edinburgh, United Kingdom (T.M.); Department of Genetics and Genomic Sciences, Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY (J.L.M.B.); Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estoni
| | - Bradford B Worrall
- From the Laboratory of Experimental Cardiology, Division Heart and Lungs (G.P., S.W.v.d.L., S.H., M.A.S., T.B., J.v.S., I.E.H., S.C.A.d.J., H.M.d.R.), Laboratory of Clinical Chemistry and Hematology, Division Laboratories and Pharmacy (G.P.), and Division Heart and Lungs, Department of Cardiology (F.W.A.), University Medical Center Utrecht, Utrecht, The Netherlands; Vascular Biology Unit, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden (H.F.A., J.L.M.B.); Institut für Schlaganfall- und Demenzforschung (ISD) Klinikum der Universität München, Munich, Germany (M.D., R.M.); Munich Cluster for Systems Neurology (SyNergy), Munich, Germany (M.D.); Department of Neurology, University of Virginia, Charlottesville (B.B.W.); Klinik für Herz- und Kreislauferkrankungen, Deutsches Herzzentrum München, Technische Universität München, Munich, Germany (H.S.); Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK) e.V., Partner Site Munich Heart Alliance, Munich, Germany (H.S.); Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom (N.J.S.); Leicester National Institute of Health Research Biomedical Research Unit in Cardiovascular Disease, Leicester, United Kingdom (N.J.S.); Department of Surgery, National University of Singapore, Singapore (D.P.V.d.K.); Cardiovascular Research Institute (CVRI), National University Health System, Singapore, Singapore (D.P.V.d.K.); Interuniversity Cardiology Institute of the Netherlands, Utrecht, The Netherlands (D.P.V.d.K.); Department of Neurosciences, University of Cambridge, Cambridge, United Kingdom (H.S.M.); The Roslin Institute, University of Edinburgh, Edinburgh, United Kingdom (T.M.); Department of Genetics and Genomic Sciences, Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY (J.L.M.B.); Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estoni
| | - Heribert Schunkert
- From the Laboratory of Experimental Cardiology, Division Heart and Lungs (G.P., S.W.v.d.L., S.H., M.A.S., T.B., J.v.S., I.E.H., S.C.A.d.J., H.M.d.R.), Laboratory of Clinical Chemistry and Hematology, Division Laboratories and Pharmacy (G.P.), and Division Heart and Lungs, Department of Cardiology (F.W.A.), University Medical Center Utrecht, Utrecht, The Netherlands; Vascular Biology Unit, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden (H.F.A., J.L.M.B.); Institut für Schlaganfall- und Demenzforschung (ISD) Klinikum der Universität München, Munich, Germany (M.D., R.M.); Munich Cluster for Systems Neurology (SyNergy), Munich, Germany (M.D.); Department of Neurology, University of Virginia, Charlottesville (B.B.W.); Klinik für Herz- und Kreislauferkrankungen, Deutsches Herzzentrum München, Technische Universität München, Munich, Germany (H.S.); Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK) e.V., Partner Site Munich Heart Alliance, Munich, Germany (H.S.); Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom (N.J.S.); Leicester National Institute of Health Research Biomedical Research Unit in Cardiovascular Disease, Leicester, United Kingdom (N.J.S.); Department of Surgery, National University of Singapore, Singapore (D.P.V.d.K.); Cardiovascular Research Institute (CVRI), National University Health System, Singapore, Singapore (D.P.V.d.K.); Interuniversity Cardiology Institute of the Netherlands, Utrecht, The Netherlands (D.P.V.d.K.); Department of Neurosciences, University of Cambridge, Cambridge, United Kingdom (H.S.M.); The Roslin Institute, University of Edinburgh, Edinburgh, United Kingdom (T.M.); Department of Genetics and Genomic Sciences, Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY (J.L.M.B.); Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estoni
| | - Nilesh J Samani
- From the Laboratory of Experimental Cardiology, Division Heart and Lungs (G.P., S.W.v.d.L., S.H., M.A.S., T.B., J.v.S., I.E.H., S.C.A.d.J., H.M.d.R.), Laboratory of Clinical Chemistry and Hematology, Division Laboratories and Pharmacy (G.P.), and Division Heart and Lungs, Department of Cardiology (F.W.A.), University Medical Center Utrecht, Utrecht, The Netherlands; Vascular Biology Unit, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden (H.F.A., J.L.M.B.); Institut für Schlaganfall- und Demenzforschung (ISD) Klinikum der Universität München, Munich, Germany (M.D., R.M.); Munich Cluster for Systems Neurology (SyNergy), Munich, Germany (M.D.); Department of Neurology, University of Virginia, Charlottesville (B.B.W.); Klinik für Herz- und Kreislauferkrankungen, Deutsches Herzzentrum München, Technische Universität München, Munich, Germany (H.S.); Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK) e.V., Partner Site Munich Heart Alliance, Munich, Germany (H.S.); Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom (N.J.S.); Leicester National Institute of Health Research Biomedical Research Unit in Cardiovascular Disease, Leicester, United Kingdom (N.J.S.); Department of Surgery, National University of Singapore, Singapore (D.P.V.d.K.); Cardiovascular Research Institute (CVRI), National University Health System, Singapore, Singapore (D.P.V.d.K.); Interuniversity Cardiology Institute of the Netherlands, Utrecht, The Netherlands (D.P.V.d.K.); Department of Neurosciences, University of Cambridge, Cambridge, United Kingdom (H.S.M.); The Roslin Institute, University of Edinburgh, Edinburgh, United Kingdom (T.M.); Department of Genetics and Genomic Sciences, Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY (J.L.M.B.); Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estoni
| | - Dominique P V de Kleijn
- From the Laboratory of Experimental Cardiology, Division Heart and Lungs (G.P., S.W.v.d.L., S.H., M.A.S., T.B., J.v.S., I.E.H., S.C.A.d.J., H.M.d.R.), Laboratory of Clinical Chemistry and Hematology, Division Laboratories and Pharmacy (G.P.), and Division Heart and Lungs, Department of Cardiology (F.W.A.), University Medical Center Utrecht, Utrecht, The Netherlands; Vascular Biology Unit, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden (H.F.A., J.L.M.B.); Institut für Schlaganfall- und Demenzforschung (ISD) Klinikum der Universität München, Munich, Germany (M.D., R.M.); Munich Cluster for Systems Neurology (SyNergy), Munich, Germany (M.D.); Department of Neurology, University of Virginia, Charlottesville (B.B.W.); Klinik für Herz- und Kreislauferkrankungen, Deutsches Herzzentrum München, Technische Universität München, Munich, Germany (H.S.); Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK) e.V., Partner Site Munich Heart Alliance, Munich, Germany (H.S.); Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom (N.J.S.); Leicester National Institute of Health Research Biomedical Research Unit in Cardiovascular Disease, Leicester, United Kingdom (N.J.S.); Department of Surgery, National University of Singapore, Singapore (D.P.V.d.K.); Cardiovascular Research Institute (CVRI), National University Health System, Singapore, Singapore (D.P.V.d.K.); Interuniversity Cardiology Institute of the Netherlands, Utrecht, The Netherlands (D.P.V.d.K.); Department of Neurosciences, University of Cambridge, Cambridge, United Kingdom (H.S.M.); The Roslin Institute, University of Edinburgh, Edinburgh, United Kingdom (T.M.); Department of Genetics and Genomic Sciences, Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY (J.L.M.B.); Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estoni
| | - Hugh S Markus
- From the Laboratory of Experimental Cardiology, Division Heart and Lungs (G.P., S.W.v.d.L., S.H., M.A.S., T.B., J.v.S., I.E.H., S.C.A.d.J., H.M.d.R.), Laboratory of Clinical Chemistry and Hematology, Division Laboratories and Pharmacy (G.P.), and Division Heart and Lungs, Department of Cardiology (F.W.A.), University Medical Center Utrecht, Utrecht, The Netherlands; Vascular Biology Unit, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden (H.F.A., J.L.M.B.); Institut für Schlaganfall- und Demenzforschung (ISD) Klinikum der Universität München, Munich, Germany (M.D., R.M.); Munich Cluster for Systems Neurology (SyNergy), Munich, Germany (M.D.); Department of Neurology, University of Virginia, Charlottesville (B.B.W.); Klinik für Herz- und Kreislauferkrankungen, Deutsches Herzzentrum München, Technische Universität München, Munich, Germany (H.S.); Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK) e.V., Partner Site Munich Heart Alliance, Munich, Germany (H.S.); Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom (N.J.S.); Leicester National Institute of Health Research Biomedical Research Unit in Cardiovascular Disease, Leicester, United Kingdom (N.J.S.); Department of Surgery, National University of Singapore, Singapore (D.P.V.d.K.); Cardiovascular Research Institute (CVRI), National University Health System, Singapore, Singapore (D.P.V.d.K.); Interuniversity Cardiology Institute of the Netherlands, Utrecht, The Netherlands (D.P.V.d.K.); Department of Neurosciences, University of Cambridge, Cambridge, United Kingdom (H.S.M.); The Roslin Institute, University of Edinburgh, Edinburgh, United Kingdom (T.M.); Department of Genetics and Genomic Sciences, Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY (J.L.M.B.); Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estoni
| | - Imo E Hoefer
- From the Laboratory of Experimental Cardiology, Division Heart and Lungs (G.P., S.W.v.d.L., S.H., M.A.S., T.B., J.v.S., I.E.H., S.C.A.d.J., H.M.d.R.), Laboratory of Clinical Chemistry and Hematology, Division Laboratories and Pharmacy (G.P.), and Division Heart and Lungs, Department of Cardiology (F.W.A.), University Medical Center Utrecht, Utrecht, The Netherlands; Vascular Biology Unit, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden (H.F.A., J.L.M.B.); Institut für Schlaganfall- und Demenzforschung (ISD) Klinikum der Universität München, Munich, Germany (M.D., R.M.); Munich Cluster for Systems Neurology (SyNergy), Munich, Germany (M.D.); Department of Neurology, University of Virginia, Charlottesville (B.B.W.); Klinik für Herz- und Kreislauferkrankungen, Deutsches Herzzentrum München, Technische Universität München, Munich, Germany (H.S.); Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK) e.V., Partner Site Munich Heart Alliance, Munich, Germany (H.S.); Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom (N.J.S.); Leicester National Institute of Health Research Biomedical Research Unit in Cardiovascular Disease, Leicester, United Kingdom (N.J.S.); Department of Surgery, National University of Singapore, Singapore (D.P.V.d.K.); Cardiovascular Research Institute (CVRI), National University Health System, Singapore, Singapore (D.P.V.d.K.); Interuniversity Cardiology Institute of the Netherlands, Utrecht, The Netherlands (D.P.V.d.K.); Department of Neurosciences, University of Cambridge, Cambridge, United Kingdom (H.S.M.); The Roslin Institute, University of Edinburgh, Edinburgh, United Kingdom (T.M.); Department of Genetics and Genomic Sciences, Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY (J.L.M.B.); Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estoni
| | - Tom Michoel
- From the Laboratory of Experimental Cardiology, Division Heart and Lungs (G.P., S.W.v.d.L., S.H., M.A.S., T.B., J.v.S., I.E.H., S.C.A.d.J., H.M.d.R.), Laboratory of Clinical Chemistry and Hematology, Division Laboratories and Pharmacy (G.P.), and Division Heart and Lungs, Department of Cardiology (F.W.A.), University Medical Center Utrecht, Utrecht, The Netherlands; Vascular Biology Unit, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden (H.F.A., J.L.M.B.); Institut für Schlaganfall- und Demenzforschung (ISD) Klinikum der Universität München, Munich, Germany (M.D., R.M.); Munich Cluster for Systems Neurology (SyNergy), Munich, Germany (M.D.); Department of Neurology, University of Virginia, Charlottesville (B.B.W.); Klinik für Herz- und Kreislauferkrankungen, Deutsches Herzzentrum München, Technische Universität München, Munich, Germany (H.S.); Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK) e.V., Partner Site Munich Heart Alliance, Munich, Germany (H.S.); Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom (N.J.S.); Leicester National Institute of Health Research Biomedical Research Unit in Cardiovascular Disease, Leicester, United Kingdom (N.J.S.); Department of Surgery, National University of Singapore, Singapore (D.P.V.d.K.); Cardiovascular Research Institute (CVRI), National University Health System, Singapore, Singapore (D.P.V.d.K.); Interuniversity Cardiology Institute of the Netherlands, Utrecht, The Netherlands (D.P.V.d.K.); Department of Neurosciences, University of Cambridge, Cambridge, United Kingdom (H.S.M.); The Roslin Institute, University of Edinburgh, Edinburgh, United Kingdom (T.M.); Department of Genetics and Genomic Sciences, Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY (J.L.M.B.); Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estoni
| | - Saskia C A de Jager
- From the Laboratory of Experimental Cardiology, Division Heart and Lungs (G.P., S.W.v.d.L., S.H., M.A.S., T.B., J.v.S., I.E.H., S.C.A.d.J., H.M.d.R.), Laboratory of Clinical Chemistry and Hematology, Division Laboratories and Pharmacy (G.P.), and Division Heart and Lungs, Department of Cardiology (F.W.A.), University Medical Center Utrecht, Utrecht, The Netherlands; Vascular Biology Unit, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden (H.F.A., J.L.M.B.); Institut für Schlaganfall- und Demenzforschung (ISD) Klinikum der Universität München, Munich, Germany (M.D., R.M.); Munich Cluster for Systems Neurology (SyNergy), Munich, Germany (M.D.); Department of Neurology, University of Virginia, Charlottesville (B.B.W.); Klinik für Herz- und Kreislauferkrankungen, Deutsches Herzzentrum München, Technische Universität München, Munich, Germany (H.S.); Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK) e.V., Partner Site Munich Heart Alliance, Munich, Germany (H.S.); Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom (N.J.S.); Leicester National Institute of Health Research Biomedical Research Unit in Cardiovascular Disease, Leicester, United Kingdom (N.J.S.); Department of Surgery, National University of Singapore, Singapore (D.P.V.d.K.); Cardiovascular Research Institute (CVRI), National University Health System, Singapore, Singapore (D.P.V.d.K.); Interuniversity Cardiology Institute of the Netherlands, Utrecht, The Netherlands (D.P.V.d.K.); Department of Neurosciences, University of Cambridge, Cambridge, United Kingdom (H.S.M.); The Roslin Institute, University of Edinburgh, Edinburgh, United Kingdom (T.M.); Department of Genetics and Genomic Sciences, Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY (J.L.M.B.); Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estoni
| | - Johan L M Björkegren
- From the Laboratory of Experimental Cardiology, Division Heart and Lungs (G.P., S.W.v.d.L., S.H., M.A.S., T.B., J.v.S., I.E.H., S.C.A.d.J., H.M.d.R.), Laboratory of Clinical Chemistry and Hematology, Division Laboratories and Pharmacy (G.P.), and Division Heart and Lungs, Department of Cardiology (F.W.A.), University Medical Center Utrecht, Utrecht, The Netherlands; Vascular Biology Unit, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden (H.F.A., J.L.M.B.); Institut für Schlaganfall- und Demenzforschung (ISD) Klinikum der Universität München, Munich, Germany (M.D., R.M.); Munich Cluster for Systems Neurology (SyNergy), Munich, Germany (M.D.); Department of Neurology, University of Virginia, Charlottesville (B.B.W.); Klinik für Herz- und Kreislauferkrankungen, Deutsches Herzzentrum München, Technische Universität München, Munich, Germany (H.S.); Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK) e.V., Partner Site Munich Heart Alliance, Munich, Germany (H.S.); Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom (N.J.S.); Leicester National Institute of Health Research Biomedical Research Unit in Cardiovascular Disease, Leicester, United Kingdom (N.J.S.); Department of Surgery, National University of Singapore, Singapore (D.P.V.d.K.); Cardiovascular Research Institute (CVRI), National University Health System, Singapore, Singapore (D.P.V.d.K.); Interuniversity Cardiology Institute of the Netherlands, Utrecht, The Netherlands (D.P.V.d.K.); Department of Neurosciences, University of Cambridge, Cambridge, United Kingdom (H.S.M.); The Roslin Institute, University of Edinburgh, Edinburgh, United Kingdom (T.M.); Department of Genetics and Genomic Sciences, Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY (J.L.M.B.); Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estoni
| | - Hester M den Ruijter
- From the Laboratory of Experimental Cardiology, Division Heart and Lungs (G.P., S.W.v.d.L., S.H., M.A.S., T.B., J.v.S., I.E.H., S.C.A.d.J., H.M.d.R.), Laboratory of Clinical Chemistry and Hematology, Division Laboratories and Pharmacy (G.P.), and Division Heart and Lungs, Department of Cardiology (F.W.A.), University Medical Center Utrecht, Utrecht, The Netherlands; Vascular Biology Unit, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden (H.F.A., J.L.M.B.); Institut für Schlaganfall- und Demenzforschung (ISD) Klinikum der Universität München, Munich, Germany (M.D., R.M.); Munich Cluster for Systems Neurology (SyNergy), Munich, Germany (M.D.); Department of Neurology, University of Virginia, Charlottesville (B.B.W.); Klinik für Herz- und Kreislauferkrankungen, Deutsches Herzzentrum München, Technische Universität München, Munich, Germany (H.S.); Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK) e.V., Partner Site Munich Heart Alliance, Munich, Germany (H.S.); Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom (N.J.S.); Leicester National Institute of Health Research Biomedical Research Unit in Cardiovascular Disease, Leicester, United Kingdom (N.J.S.); Department of Surgery, National University of Singapore, Singapore (D.P.V.d.K.); Cardiovascular Research Institute (CVRI), National University Health System, Singapore, Singapore (D.P.V.d.K.); Interuniversity Cardiology Institute of the Netherlands, Utrecht, The Netherlands (D.P.V.d.K.); Department of Neurosciences, University of Cambridge, Cambridge, United Kingdom (H.S.M.); The Roslin Institute, University of Edinburgh, Edinburgh, United Kingdom (T.M.); Department of Genetics and Genomic Sciences, Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY (J.L.M.B.); Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estoni
| | - Folkert W Asselbergs
- From the Laboratory of Experimental Cardiology, Division Heart and Lungs (G.P., S.W.v.d.L., S.H., M.A.S., T.B., J.v.S., I.E.H., S.C.A.d.J., H.M.d.R.), Laboratory of Clinical Chemistry and Hematology, Division Laboratories and Pharmacy (G.P.), and Division Heart and Lungs, Department of Cardiology (F.W.A.), University Medical Center Utrecht, Utrecht, The Netherlands; Vascular Biology Unit, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden (H.F.A., J.L.M.B.); Institut für Schlaganfall- und Demenzforschung (ISD) Klinikum der Universität München, Munich, Germany (M.D., R.M.); Munich Cluster for Systems Neurology (SyNergy), Munich, Germany (M.D.); Department of Neurology, University of Virginia, Charlottesville (B.B.W.); Klinik für Herz- und Kreislauferkrankungen, Deutsches Herzzentrum München, Technische Universität München, Munich, Germany (H.S.); Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK) e.V., Partner Site Munich Heart Alliance, Munich, Germany (H.S.); Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom (N.J.S.); Leicester National Institute of Health Research Biomedical Research Unit in Cardiovascular Disease, Leicester, United Kingdom (N.J.S.); Department of Surgery, National University of Singapore, Singapore (D.P.V.d.K.); Cardiovascular Research Institute (CVRI), National University Health System, Singapore, Singapore (D.P.V.d.K.); Interuniversity Cardiology Institute of the Netherlands, Utrecht, The Netherlands (D.P.V.d.K.); Department of Neurosciences, University of Cambridge, Cambridge, United Kingdom (H.S.M.); The Roslin Institute, University of Edinburgh, Edinburgh, United Kingdom (T.M.); Department of Genetics and Genomic Sciences, Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY (J.L.M.B.); Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estoni
| |
Collapse
|
309
|
Baird A, Costantini T, Coimbra R, Eliceiri BP. Injury, inflammation and the emergence of human-specific genes. Wound Repair Regen 2016; 24:602-6. [PMID: 26874655 PMCID: PMC5021143 DOI: 10.1111/wrr.12422] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Accepted: 02/08/2016] [Indexed: 12/22/2022]
Abstract
In light of the central role of inflammation in normal wound repair and regeneration, we hypothesize that the preponderance of human‐specific genes expressed in human inflammatory cells is commensurate with the genetic versatility of inflammatory response and the emergence of injuries associated with uniquely hominid behaviors, like a bipedal posture and the use of tools, weapons and fire. The hypothesis underscores the need to study human‐specific signaling pathways in experimental models of injury and infers that a selection of human‐specific genes, driven in part by the response to injury, may have facilitated the emergence of multifunctional genes expressed in other tissues.
Collapse
Affiliation(s)
- Andrew Baird
- Division of Trauma, Surgical Critical Care, Burns and Acute Care Surgery, Department of Surgery, University of California San Diego School of Medicine, La Jolla, San Diego, California
| | - Todd Costantini
- Division of Trauma, Surgical Critical Care, Burns and Acute Care Surgery, Department of Surgery, University of California San Diego School of Medicine, La Jolla, San Diego, California
| | - Raul Coimbra
- Division of Trauma, Surgical Critical Care, Burns and Acute Care Surgery, Department of Surgery, University of California San Diego School of Medicine, La Jolla, San Diego, California
| | - Brian P Eliceiri
- Division of Trauma, Surgical Critical Care, Burns and Acute Care Surgery, Department of Surgery, University of California San Diego School of Medicine, La Jolla, San Diego, California
| |
Collapse
|
310
|
Haffner-Luntzer M, Kovtun A, Rapp AE, Ignatius A. Mouse Models in Bone Fracture Healing Research. ACTA ACUST UNITED AC 2016. [DOI: 10.1007/s40610-016-0037-3] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
|
311
|
Chao W, Belmonte C, Benitez del Castillo JM, Bron AJ, Dua HS, Nichols KK, Novack GD, Schrader S, Willcox MD, Wolffsohn JS, Sullivan DA. Report of the Inaugural Meeting of the TFOS i2 = initiating innovation Series: Targeting the Unmet Need for Dry Eye Treatment. Ocul Surf 2016; 14:264-316. [DOI: 10.1016/j.jtos.2015.11.003] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Revised: 11/09/2015] [Accepted: 11/11/2015] [Indexed: 01/09/2023]
|
312
|
Abstract
Progress in magnetic resonance imaging (MRI) now makes it possible to identify the major white matter tracts in the living human brain. These tracts are important because they carry many of the signals communicated between different brain regions. MRI methods coupled with biophysical modeling can measure the tissue properties and structural features of the tracts that impact our ability to think, feel, and perceive. This review describes the fundamental ideas of the MRI methods used to identify the major white matter tracts in the living human brain.
Collapse
Affiliation(s)
- Brian A Wandell
- Department of Psychology and Stanford Neurosciences Institute, Stanford University, Stanford, California 94305;
| |
Collapse
|
313
|
Nielsen BW, Bonney EA, Pearce BD, Donahue LR, Sarkar IN. A Cross-Species Analysis of Animal Models for the Investigation of Preterm Birth Mechanisms. Reprod Sci 2016; 23:482-91. [PMID: 26377998 PMCID: PMC5933186 DOI: 10.1177/1933719115604729] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND Spontaneous preterm birth is the leading cause of neonatal morbidity and mortality worldwide. The ability to examine the exact mechanisms underlying this syndrome in humans is limited. Therefore, the study of animal models is critical to unraveling the key physiologic mechanisms that control the timing of birth. The purpose of this review is to facilitate enhanced assimilation of the literature on animal models of preterm birth by a broad range of investigators. METHODS Using classical systematic and informatics search techniques of the available literature through 2012, a database of intact animal models was generated. Research librarians generated a list of articles using multiple databases. From these articles, a comprehensive list of Medical Subject Headings (MeSH) was created. Using mathematical modeling, significant MeSH descriptors were determined, and a MEDLINE search algorithm was created. The articles were reviewed for mechanism of labor induction categorized by species. RESULTS Existing animal models of preterm birth comprise specific interventions to induce preterm birth, as no animal model was identified that exhibits natural spontaneous preterm birth at an incidence comparable to that of the humans. A search algorithm was developed which when used results in a comprehensive list of agents used to induce preterm delivery in a host of animal species. The evolution of 3 specific animal models--sheep, mice, and rats--has demonstrated a clear shift in focus in the literature from endocrine to inflammatory agents of preterm birth induction. CONCLUSION The process of developing a search algorithm to provide efficient access to information on animal models of preterm birth illustrates the need for a more precise organization of the literature to allow the investigator to focus on distinctly maternal versus fetal outcomes.
Collapse
Affiliation(s)
- Brian W Nielsen
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Vermont College of Medicine, Burlington, VT, USA
| | - Elizabeth A Bonney
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Vermont College of Medicine, Burlington, VT, USA
| | - Bradley D Pearce
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | | | - Indra Neil Sarkar
- Center for Biomedical Informatics, Warren Alpert Medical School of Brown University, Providence, RI, USA
| |
Collapse
|
314
|
Hordijk PL. Recent insights into endothelial control of leukocyte extravasation. Cell Mol Life Sci 2016; 73:1591-608. [PMID: 26794844 PMCID: PMC11108429 DOI: 10.1007/s00018-016-2136-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Revised: 01/07/2016] [Accepted: 01/11/2016] [Indexed: 12/30/2022]
Abstract
In the process of leukocyte migration from the circulation across the vascular wall, the crosstalk with endothelial cells that line the blood vessels is essential. It is now firmly established that in endothelial cells important signaling events are initiated upon leukocyte adhesion that impinge on the regulation of cell-cell contact and control the efficiency of transendothelial migration. In addition, several external factors such as shear force and vascular stiffness were recently identified as important regulators of endothelial signaling and, consequently, leukocyte transmigration. Here, I review recent insights into endothelial signaling events that are linked to leukocyte migration across the vessel wall. In this field, protein phosphorylation and Rho-mediated cytoskeletal dynamics are still widely studied using increasingly sophisticated mouse models. In addition, activation of tyrosine phosphatases, changes in endothelial cell stiffness as well as different vascular beds have all been established as important factors in endothelial signaling and leukocyte transmigration. Finally, I address less-well-studied but interesting components in the endothelium that also control transendothelial migration, such as the ephrins and their Eph receptors, that provide novel insights in the complexity associated with this process.
Collapse
Affiliation(s)
- Peter L Hordijk
- Department of Molecular Cell Biology, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, Swammerdam Institute for Life Sciences, University of Amsterdam, Plesmanlaan 125, 1066 CX, Amsterdam, The Netherlands.
- Department of Physiology, VU University Medical Center, Amsterdam, The Netherlands.
| |
Collapse
|
315
|
Djoumerska-Alexieva I, Roumenina L, Pashov A, Dimitrov J, Hadzhieva M, Lindig S, Voynova E, Dimitrova P, Ivanovska N, Bockmeyer C, Stefanova Z, Fitting C, Bläss M, Claus R, von Gunten S, Kaveri S, Cavaillon JM, Bauer M, Vassilev T. Intravenous Immunoglobulin with Enhanced Polyspecificity Improves Survival in Experimental Sepsis and Aseptic Systemic Inflammatory Response Syndromes. Mol Med 2016; 21:1002-1010. [PMID: 26701312 DOI: 10.2119/molmed.2014.00224] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Accepted: 12/14/2015] [Indexed: 11/06/2022] Open
Abstract
Sepsis is a major cause for death worldwide. Numerous interventional trials with agents neutralizing single proinflammatory mediators have failed to improve survival in sepsis and aseptic systemic inflammatory response syndromes. This failure could be explained by the widespread gene expression dysregulation known as "genomic storm" in these patients. A multifunctional polyspecific therapeutic agent might be needed to thwart the effects of this storm. Licensed pooled intravenous immunoglobulin preparations seemed to be a promising candidate, but they have also failed in their present form to prevent sepsis-related death. We report here the protective effect of a single dose of intravenous immunoglobulin preparations with additionally enhanced polyspecificity in three models of sepsis and aseptic systemic inflammation. The modification of the pooled immunoglobulin G molecules by exposure to ferrous ions resulted in their newly acquired ability to bind some proinflammatory molecules, complement components and endogenous "danger" signals. The improved survival in endotoxemia was associated with serum levels of proinflammatory cytokines, diminished complement consumption and normalization of the coagulation time. We suggest that intravenous immunoglobulin preparations with additionally enhanced polyspecificity have a clinical potential in sepsis and related systemic inflammatory syndromes.
Collapse
Affiliation(s)
- Iglika Djoumerska-Alexieva
- Department of Immunology, Stefan Angelov Institute of Microbiology, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Lubka Roumenina
- INSERM UMRS 1138, Centre de Recherche des Cordeliers, Paris, France
| | - Anastas Pashov
- Department of Immunology, Stefan Angelov Institute of Microbiology, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Jordan Dimitrov
- Department of Immunology, Stefan Angelov Institute of Microbiology, Bulgarian Academy of Sciences, Sofia, Bulgaria.,INSERM UMRS 1138, Centre de Recherche des Cordeliers, Paris, France
| | - Maya Hadzhieva
- Department of Immunology, Stefan Angelov Institute of Microbiology, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Sandro Lindig
- Center for Sepsis Control and Care, University Hospital, Friedrich Schiller University, Jena, Germany
| | - Elisaveta Voynova
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Petya Dimitrova
- Department of Immunology, Stefan Angelov Institute of Microbiology, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Nina Ivanovska
- Department of Immunology, Stefan Angelov Institute of Microbiology, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | | | - Zvetanka Stefanova
- Department of Immunology, Stefan Angelov Institute of Microbiology, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Catherine Fitting
- Cytokines and Inflammation Unit, Institut Pasteur, Paris, France; and
| | - Markus Bläss
- Center for Sepsis Control and Care, University Hospital, Friedrich Schiller University, Jena, Germany
| | - Ralf Claus
- Center for Sepsis Control and Care, University Hospital, Friedrich Schiller University, Jena, Germany
| | | | - Srini Kaveri
- INSERM UMRS 1138, Centre de Recherche des Cordeliers, Paris, France
| | | | - Michael Bauer
- Center for Sepsis Control and Care, University Hospital, Friedrich Schiller University, Jena, Germany
| | - Tchavdar Vassilev
- Department of Immunology, Stefan Angelov Institute of Microbiology, Bulgarian Academy of Sciences, Sofia, Bulgaria.,Center for Sepsis Control and Care, University Hospital, Friedrich Schiller University, Jena, Germany
| |
Collapse
|
316
|
Fullerton JN, Gilroy DW. Resolution of inflammation: a new therapeutic frontier. Nat Rev Drug Discov 2016; 15:551-67. [PMID: 27020098 DOI: 10.1038/nrd.2016.39] [Citation(s) in RCA: 638] [Impact Index Per Article: 70.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Dysregulated inflammation is a central pathological process in diverse disease states. Traditionally, therapeutic approaches have sought to modulate the pro- or anti-inflammatory limbs of inflammation, with mixed success. However, insight into the pathways by which inflammation is resolved has highlighted novel opportunities to pharmacologically manipulate these processes - a strategy that might represent a complementary (and perhaps even superior) therapeutic approach. This Review discusses the state of the art in the biology of resolution of inflammation, highlighting the opportunities and challenges for translational research in this field.
Collapse
Affiliation(s)
- James N Fullerton
- Centre for Clinical Pharmacology and Therapeutics, Division of Medicine, 5 University Street, University College London, London WC1E 6JJ, UK
| | - Derek W Gilroy
- Centre for Clinical Pharmacology and Therapeutics, Division of Medicine, 5 University Street, University College London, London WC1E 6JJ, UK
| |
Collapse
|
317
|
Castro NP, Fedorova-Abrams ND, Merchant AS, Rangel MC, Nagaoka T, Karasawa H, Klauzinska M, Hewitt SM, Biswas K, Sharan SK, Salomon DS. Cripto-1 as a novel therapeutic target for triple negative breast cancer. Oncotarget 2016; 6:11910-29. [PMID: 26059540 PMCID: PMC4494913 DOI: 10.18632/oncotarget.4182] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Accepted: 05/09/2015] [Indexed: 12/21/2022] Open
Abstract
Triple-negative breast cancer (TNBC) presents the poorest prognosis among the breast cancer subtypes and no current standard therapy. Here, we performed an in-depth molecular analysis of a mouse model that establishes spontaneous lung metastasis from JygMC(A) cells. These primary tumors resembled the triple-negative breast cancer (TNBC) both phenotypically and molecularly. Morphologically, primary tumors presented both epithelial and spindle-like cells but displayed only adenocarcinoma-like features in lung parenchyma. The use of laser-capture microdissection combined with Nanostring mRNA and microRNA analysis revealed overexpression of either epithelial and miRNA-200 family or mesenchymal markers in adenocarcinoma and mesenchymal regions, respectively. Cripto-1, an embryonic stem cell marker, was present in spindle-like areas and its promoter showed activity in primary tumors. Cripto-1 knockout by the CRISPR-Cas9 system inhibited tumor growth and pulmonary metastasis. Our findings show characterization of a novel mouse model that mimics the TNBC and reveal Cripto-1 as a TNBC target hence may offer alternative treatment strategies for TNBC.
Collapse
Affiliation(s)
- Nadia P Castro
- Tumor Growth Factor Section, Mouse Cancer Genetics Program, National Cancer Institute, Frederick, MD, USA
| | | | - Anand S Merchant
- CCRIFX Bioinformatics Core, National Cancer Institute, Bethesda, MD, USA
| | - Maria Cristina Rangel
- Tumor Growth Factor Section, Mouse Cancer Genetics Program, National Cancer Institute, Frederick, MD, USA
| | - Tadahiro Nagaoka
- Tumor Growth Factor Section, Mouse Cancer Genetics Program, National Cancer Institute, Frederick, MD, USA
| | - Hideaki Karasawa
- Tumor Growth Factor Section, Mouse Cancer Genetics Program, National Cancer Institute, Frederick, MD, USA
| | - Malgorzata Klauzinska
- Tumor Growth Factor Section, Mouse Cancer Genetics Program, National Cancer Institute, Frederick, MD, USA
| | - Stephen M Hewitt
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Kajal Biswas
- Genetics of Cancer Susceptibility Section, Mouse Cancer Genetics Program, National Cancer Institute, Frederick, MD, USA
| | - Shyam K Sharan
- Genetics of Cancer Susceptibility Section, Mouse Cancer Genetics Program, National Cancer Institute, Frederick, MD, USA
| | - David S Salomon
- Tumor Growth Factor Section, Mouse Cancer Genetics Program, National Cancer Institute, Frederick, MD, USA
| |
Collapse
|
318
|
Barber AE, Fleming BA, Mulvey MA. Similarly Lethal Strains of Extraintestinal Pathogenic Escherichia coli Trigger Markedly Diverse Host Responses in a Zebrafish Model of Sepsis. mSphere 2016; 1:e00062-16. [PMID: 27303721 PMCID: PMC4894679 DOI: 10.1128/msphere.00062-16] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Accepted: 03/29/2016] [Indexed: 02/06/2023] Open
Abstract
In individuals with sepsis, the infecting microbes are commonly viewed as generic inducers of inflammation while the host background is considered the primary variable affecting disease progression and outcome. To study the effects of bacterial strain differences on the maladaptive immune responses that are induced during sepsis, we employed a novel zebrafish embryo infection model using extraintestinal pathogenic Escherichia coli (ExPEC) isolates. These genetically diverse pathogens are a leading cause of sepsis and are becoming increasingly dangerous because of the rise of multidrug-resistant strains. Zebrafish infected with ExPEC isolates exhibit many of the pathophysiological features seen in septic human patients, including dysregulated inflammatory responses (cytokine storms), tachycardia, endothelial leakage, and progressive edema. However, only a limited subset of ExPEC isolates can trigger a sepsis-like state and death of the host when introduced into the bloodstream. Mirroring the situation in human patients, antibiotic therapy reduced ExPEC titers and improved host survival rates but was only effective within limited time frames that varied, depending on the infecting pathogen. Intriguingly, we find that phylogenetically distant but similarly lethal ExPEC isolates can stimulate markedly different host transcriptional responses, including disparate levels of inflammatory mediators. These differences correlate with the amounts of bacterial flagellin expression during infection, as well as differential activation of Toll-like receptor 5 by discrete flagellar serotypes. Altogether, this work establishes zebrafish as a relevant model of key aspects of human sepsis and highlights the ability of genetically distinct ExPEC isolates to induce divergent host responses independently of baseline host attributes. IMPORTANCE Sepsis is a life-threatening systemic inflammatory condition that is initiated by the presence of microorganisms in the bloodstream. In the United States, sepsis due to ExPEC and other pathogens kills well over a quarter of a million people each year and is associated with tremendous health care costs. A high degree of heterogeneity in the signs and symptomology of sepsis makes this disease notoriously difficult to effectively diagnose and manage. Here, using a zebrafish model of sepsis, we find that similarly lethal but genetically distinct ExPEC isolates can elicit notably disparate host responses. These variances are in part due to differences in the levels and types of flagellin that are expressed by the infecting ExPEC strains. A better understanding of the variable impact that bacterial factors like flagellin have on host responses during sepsis could lead to improved diagnostic and therapeutic approaches to these often deadly infections. Podcast: A podcast concerning this article is available.
Collapse
Affiliation(s)
- Amelia E Barber
- Division of Microbiology and Immunology, Pathology Department, University of Utah, Salt Lake City, Utah, USA
| | - Brittany A Fleming
- Division of Microbiology and Immunology, Pathology Department, University of Utah, Salt Lake City, Utah, USA
| | - Matthew A Mulvey
- Division of Microbiology and Immunology, Pathology Department, University of Utah, Salt Lake City, Utah, USA
| |
Collapse
|
319
|
Gröger M, Rennert K, Giszas B, Weiß E, Dinger J, Funke H, Kiehntopf M, Peters FT, Lupp A, Bauer M, Claus RA, Huber O, Mosig AS. Monocyte-induced recovery of inflammation-associated hepatocellular dysfunction in a biochip-based human liver model. Sci Rep 2016; 6:21868. [PMID: 26902749 PMCID: PMC4763209 DOI: 10.1038/srep21868] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 02/02/2016] [Indexed: 12/19/2022] Open
Abstract
Liver dysfunction is an early event in sepsis-related multi-organ failure. We here report the establishment and characterization of a microfluidically supported in vitro organoid model of the human liver sinusoid. The liver organoid is composed of vascular and hepatocyte cell layers integrating non-parenchymal cells closely reflecting tissue architecture and enables physiological cross-communication in a bio-inspired fashion. Inflammation-associated liver dysfunction was mimicked by stimulation with various agonists of toll-like receptors. TLR-stimulation induced the release of pro- and anti-inflammatory cytokines and diminished expression of endothelial VE-cadherin, hepatic MRP-2 transporter and apolipoprotein B (ApoB), resulting in an inflammation-related endothelial barrier disruption and hepatocellular dysfunction in the liver organoid. However, interaction of the liver organoid with human monocytes attenuated inflammation-related cell responses and restored MRP-2 transporter activity, ApoB expression and albumin/urea production. The cellular events observed in the liver organoid closely resembled pathophysiological responses in the well-established sepsis model of peritoneal contamination and infection (PCI) in mice and clinical observations in human sepsis. We therefore conclude that this human liver organoid model is a valuable tool to investigate sepsis-related liver dysfunction and subsequent immune cell-related tissue repair/remodeling processes.
Collapse
Affiliation(s)
- Marko Gröger
- Institute of Biochemistry II, Jena University Hospital, 07743 Jena, Germany.,Center for Sepsis Control and Care, Jena University Hospital, Jena, 07747 Jena, Germany
| | - Knut Rennert
- Institute of Biochemistry II, Jena University Hospital, 07743 Jena, Germany.,Center for Sepsis Control and Care, Jena University Hospital, Jena, 07747 Jena, Germany
| | - Benjamin Giszas
- Department of Anesthesiology and Intensive Care, Jena University Hospital, Jena 07747 Jena, Germany.,Center for Sepsis Control and Care, Jena University Hospital, Jena, 07747 Jena, Germany
| | - Elisabeth Weiß
- Institute of Biochemistry II, Jena University Hospital, 07743 Jena, Germany
| | - Julia Dinger
- Institute of Forensic Medicine, Jena University Hospital, 07743 Jena, Germany
| | - Harald Funke
- Molecular Hemostaseology, Jena University Hospital, Jena, 07747 Jena, Germany
| | - Michael Kiehntopf
- Institute of Clinical Chemistry and Laboratory Diagnostics, Jena University Hospital, 07747 Jena, Germany.,Center for Sepsis Control and Care, Jena University Hospital, Jena, 07747 Jena, Germany
| | - Frank T Peters
- Institute of Forensic Medicine, Jena University Hospital, 07743 Jena, Germany
| | - Amelie Lupp
- Institute of Pharmacology and Toxicology, Jena University Hospital, Jena, Germany
| | - Michael Bauer
- Department of Anesthesiology and Intensive Care, Jena University Hospital, Jena 07747 Jena, Germany.,Center for Sepsis Control and Care, Jena University Hospital, Jena, 07747 Jena, Germany
| | - Ralf A Claus
- Department of Anesthesiology and Intensive Care, Jena University Hospital, Jena 07747 Jena, Germany.,Center for Sepsis Control and Care, Jena University Hospital, Jena, 07747 Jena, Germany
| | - Otmar Huber
- Institute of Biochemistry II, Jena University Hospital, 07743 Jena, Germany.,Center for Sepsis Control and Care, Jena University Hospital, Jena, 07747 Jena, Germany
| | - Alexander S Mosig
- Institute of Biochemistry II, Jena University Hospital, 07743 Jena, Germany.,Center for Sepsis Control and Care, Jena University Hospital, Jena, 07747 Jena, Germany
| |
Collapse
|
320
|
Involvement of B cells in non-infectious uveitis. Clin Transl Immunology 2016; 5:e63. [PMID: 26962453 PMCID: PMC4771944 DOI: 10.1038/cti.2016.2] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2015] [Revised: 01/04/2016] [Accepted: 01/04/2016] [Indexed: 12/14/2022] Open
Abstract
Non-infectious uveitis-or intraocular inflammatory disease-causes substantial visual morbidity and reduced quality of life amongst affected individuals. To date, research of pathogenic mechanisms has largely been focused on processes involving T lymphocyte and/or myeloid leukocyte populations. Involvement of B lymphocytes has received relatively little attention. In contrast, B-cell pathobiology is a major field within general immunological research, and large clinical trials have showed that treatments targeting B cells are highly effective for multiple systemic inflammatory diseases. B cells, including the terminally differentiated plasma cell that produces antibody, are found in the human eye in different forms of non-infectious uveitis; in some cases, these cells outnumber other leukocyte subsets. Recent case reports and small case series suggest that B-cell blockade may be therapeutic for patients with non-infectious uveitis. As well as secretion of antibody, B cells may promote intraocular inflammation by presentation of antigen to T cells, production of multiple inflammatory cytokines and support of T-cell survival. B cells may also perform various immunomodulatory activities within the eye. This translational review summarizes the evidence for B-cell involvement in non-infectious uveitis, and considers the potential contributions of B cells to the development and control of the disease. Manipulations of B cells and/or their products are promising new approaches to the treatment of non-infectious uveitis.
Collapse
|
321
|
Rasid O, Cavaillon JM. Recent developments in severe sepsis research: from bench to bedside and back. Future Microbiol 2016; 11:293-314. [PMID: 26849633 DOI: 10.2217/fmb.15.133] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Severe sepsis remains a worldwide threat, not only in industrialized countries, due to their aging population, but also in developing countries where there still are numerous cases of neonatal and puerperal sepsis. Tools for early diagnosis, a prerequisite for rapid and appropriate antibiotic therapy, are still required. In this review, we highlight some recent developments in our understanding of the associated systemic inflammatory response that help deciphering pathophysiology (e.g., epigenetic, miRNA, regulatory loops, compartmentalization, apoptosis and synergy) and discuss some of the consequences of sepsis (e.g., immune status, neurological and muscular alterations). We also emphasize the challenge to better define animal models and discuss past failures in clinical investigations in order to define new efficient therapies.
Collapse
Affiliation(s)
- Orhan Rasid
- Unit Cytokines & Inflammation, Institut Pasteur, 28 rue Dr. Roux, Paris, France
| | - Jean-Marc Cavaillon
- Unit Cytokines & Inflammation, Institut Pasteur, 28 rue Dr. Roux, Paris, France
| |
Collapse
|
322
|
Justice MJ, Dhillon P. Using the mouse to model human disease: increasing validity and reproducibility. Dis Model Mech 2016; 9:101-3. [PMID: 26839397 PMCID: PMC4770152 DOI: 10.1242/dmm.024547] [Citation(s) in RCA: 191] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Experiments that use the mouse as a model for disease have recently come under scrutiny because of the repeated failure of data, particularly derived from preclinical studies, to be replicated or translated to humans. The usefulness of mouse models has been questioned because of irreproducibility and poor recapitulation of human conditions. Newer studies, however, point to bias in reporting results and improper data analysis as key factors that limit reproducibility and validity of preclinical mouse research. Inaccurate and incomplete descriptions of experimental conditions also contribute. Here, we provide guidance on best practice in mouse experimentation, focusing on appropriate selection and validation of the model, sources of variation and their influence on phenotypic outcomes, minimum requirements for control sets, and the importance of rigorous statistics. Our goal is to raise the standards in mouse disease modeling to enhance reproducibility, reliability and clinical translation of findings.
Collapse
Affiliation(s)
- Monica J Justice
- Hospital for Sick Children, The Peter Gilgan Centre for Research and Learning, SickKids Research Institute, 686 Bay St, 14.9716, Toronto, Ontario, Canada M5G 0A4
| | - Paraminder Dhillon
- Disease Models & Mechanisms, The Company of Biologists, Bidder Building, Station Road, Histon, Cambridge CB24 9LF, UK
| |
Collapse
|
323
|
Gharib SA, Mar D, Bomsztyk K, Denisenko O, Dhanireddy S, Liles WC, Altemeier WA. SYSTEM-WIDE MAPPING OF ACTIVATED CIRCUITRY IN EXPERIMENTAL SYSTEMIC INFLAMMATORY RESPONSE SYNDROME. Shock 2016; 45:148-56. [PMID: 26536201 PMCID: PMC4715796 DOI: 10.1097/shk.0000000000000507] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Sepsis-induced multiple organ dysfunction syndrome (MODS) is a major cause of morbidity and mortality in critically ill patients and remains impervious to most therapeutic interventions. We utilized a clinically relevant murine model of systemic inflammatory response syndrome (SIRS) during early MODS induced by ventilator-associated pneumonia to systematically delineate pathways dysregulated in lung, liver, and kidney. We focused on processes commonly activated across at-risk organs and constructed an SIRS-associated network based on connectivity among the gene members of these functionally coherent pathways. Our analyses led to the identification of several putative drivers of early MODS whose expression was regulated by epidermal growth factor receptor. Our unbiased, integrative method is a promising approach to unravel mechanisms in system-wide disorders afflicting multiple compartments such as sepsis-induced MODS, and identify putative therapeutic targets.
Collapse
Affiliation(s)
- Sina A. Gharib
- Computational Medicine Core, University of Washington, Seattle WA
- Center for Lung Biology, University of Washington, Seattle WA
- Division of Pulmonary and Critical Care Medicine, University of Washington, Seattle WA
- Department of Medicine, University of Washington, Seattle WA
| | - Daniel Mar
- Department of Medicine, University of Washington, Seattle WA
| | - Karol Bomsztyk
- Department of Medicine, University of Washington, Seattle WA
| | - Oleg Denisenko
- Department of Medicine, University of Washington, Seattle WA
| | | | - W. Conrad Liles
- Center for Lung Biology, University of Washington, Seattle WA
- Department of Medicine, University of Washington, Seattle WA
| | - William A. Altemeier
- Center for Lung Biology, University of Washington, Seattle WA
- Division of Pulmonary and Critical Care Medicine, University of Washington, Seattle WA
- Department of Medicine, University of Washington, Seattle WA
| |
Collapse
|
324
|
Tomato Aqueous Extract Modulates the Inflammatory Profile of Immune Cells and Endothelial Cells. Molecules 2016; 21:168. [PMID: 26840280 PMCID: PMC6273921 DOI: 10.3390/molecules21020168] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Revised: 01/13/2016] [Accepted: 01/20/2016] [Indexed: 01/18/2023] Open
Abstract
Nutrients transiently or chronically modulate functional and biochemical characteristics of cells and tissues both in vivo and in vitro. The influence of tomato aqueous extract (TAE) on the in vitro inflammatory response of activated human peripheral blood leukocytes (PBLs) and macrophages was investigated. Its effect on endothelial dysfunction (ED) was analyzed in human umbilical vein endothelial cells (HUVECs). Murine macrophages (RAW264.7 cells), PBLs and HUVECs were incubated with TAE. They were activated with LPS or TNF-α in order to induce inflammatory processes and ED, respectively. Inflammatory mediators and adhesion molecules were measured by immune assay-based multiplex analysis. Gene expression was quantified by RT-PCR. TAE altered the production of interleukins (IL-1β, IL-6, IL-10, IL-12) and chemokines (CCL2/MCP-1, CCL3/MIP-1α, CCL5/RANTES, CXCL8/IL-8, CXCL10/IP-10) in PBLs. TAE reduced ED-associated expression of adhesion molecules (ICAM-1, VCAM-1) in endothelial cell. In macrophages, the production of nitric oxide, PGE2, cytokines and ILs (TNF-α, IL-1β, IL-6, IL-12), which reflects chronic inflammatory processes, was reduced. Adenosine was identified as the main bioactive of TAE. Thus, TAE had cell-specific and context-dependent effects. We infer from these in vitro data, that during acute inflammation TAE enhances cellular alertness and therefore the sensing of disturbed immune homeostasis in the vascular-endothelial compartment. Conversely, it blunts inflammatory mediators in macrophages during chronic inflammation. A novel concept of immune regulation by this extract is proposed.
Collapse
|
325
|
Ariffin JK, Kapetanovic R, Schaale K, Gatica-Andrades M, Blumenthal A, Schroder K, Sweet MJ. The E3 ubiquitin ligase RNF144B is LPS-inducible in human, but not mouse, macrophages and promotes inducible IL-1β expression. J Leukoc Biol 2016; 100:155-61. [PMID: 26819317 DOI: 10.1189/jlb.2ab0815-339r] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Accepted: 01/04/2016] [Indexed: 11/24/2022] Open
Abstract
Differences in human and mouse immune responses may partly reflect species-specific adaptations and can provide important insights into human immunity. In this study, we show that RNF144B, which encodes an E3 ubiquitin ligase, was lipopolysaccharide-inducible in primary human macrophages and in human macrophage-like THP-1 cells. In contrast, Rnf144b was not lipopolysaccharide-inducible in several mouse cell populations, including primary macrophages from C57BL/6 and BALB/c mice and RAW264.7 macrophages. Similarly, Rnf144b was not up-regulated by infection of C57BL/6 mice with Escherichia coli Although the human and mouse RNF144B genes have conserved transcription start sites, cap analysis of gene expression data confirmed that the RNF144B promoter directs transcription in human but not mouse macrophages. The human and mouse RNF144B genes are controlled by highly conserved TATA-containing promoters, but subtle differences in transcription factor binding sites may account for differential regulation. Using gene silencing, we showed that RNF144B is necessary for priming of inflammasome responses in primary human macrophages. Specifically, RNF144B promotes lipopolysaccharide-inducible IL-1b mRNA expression but does not regulate expression of several other lipopolysaccharide-inducible cytokines (e.g., interleukin-10, interferon-γ) or affect expression of inflammasome components or substrates (e.g., procaspase-1, pro-interleukin-18). Our findings thus revealed a species-specific regulatory mechanism for selective inflammasome priming in human macrophages.
Collapse
Affiliation(s)
- Juliana K Ariffin
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, Queensland, Australia; IMB Centre for Inflammation and Disease Research, The University of Queensland, Brisbane, Queensland, Australia; Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, Queensland, Australia; and
| | - Ronan Kapetanovic
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, Queensland, Australia; IMB Centre for Inflammation and Disease Research, The University of Queensland, Brisbane, Queensland, Australia; Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, Queensland, Australia; and
| | - Kolja Schaale
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, Queensland, Australia; IMB Centre for Inflammation and Disease Research, The University of Queensland, Brisbane, Queensland, Australia; Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, Queensland, Australia; and
| | - Marcela Gatica-Andrades
- Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, Queensland, Australia; and The University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| | - Antje Blumenthal
- Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, Queensland, Australia; and The University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| | - Kate Schroder
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, Queensland, Australia; IMB Centre for Inflammation and Disease Research, The University of Queensland, Brisbane, Queensland, Australia; Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, Queensland, Australia; and
| | - Matthew J Sweet
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, Queensland, Australia; IMB Centre for Inflammation and Disease Research, The University of Queensland, Brisbane, Queensland, Australia; Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, Queensland, Australia; and
| |
Collapse
|
326
|
Godec J, Tan Y, Liberzon A, Tamayo P, Bhattacharya S, Butte AJ, Mesirov JP, Haining WN. Compendium of Immune Signatures Identifies Conserved and Species-Specific Biology in Response to Inflammation. Immunity 2016; 44:194-206. [PMID: 26795250 PMCID: PMC5330663 DOI: 10.1016/j.immuni.2015.12.006] [Citation(s) in RCA: 208] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Revised: 08/02/2015] [Accepted: 09/30/2015] [Indexed: 01/07/2023]
Abstract
Gene-expression profiling has become a mainstay in immunology, but subtle changes in gene networks related to biological processes are hard to discern when comparing various datasets. For instance, conservation of the transcriptional response to sepsis in mouse models and human disease remains controversial. To improve transcriptional analysis in immunology, we created ImmuneSigDB: a manually annotated compendium of ∼5,000 gene-sets from diverse cell states, experimental manipulations, and genetic perturbations in immunology. Analysis using ImmuneSigDB identified signatures induced in activated myeloid cells and differentiating lymphocytes that were highly conserved between humans and mice. Sepsis triggered conserved patterns of gene expression in humans and mouse models. However, we also identified species-specific biological processes in the sepsis transcriptional response: although both species upregulated phagocytosis-related genes, a mitosis signature was specific to humans. ImmuneSigDB enables granular analysis of transcriptomic data to improve biological understanding of immune processes of the human and mouse immune systems.
Collapse
Affiliation(s)
- Jernej Godec
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA; Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Yan Tan
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Bioinformatics Program, Boston University, Boston, MA 02215, USA
| | - Arthur Liberzon
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Pablo Tamayo
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Sanchita Bhattacharya
- Institute for Computational Health Science, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Atul J Butte
- Institute for Computational Health Science, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Jill P Mesirov
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Bioinformatics Program, Boston University, Boston, MA 02215, USA
| | - W Nicholas Haining
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Division of Hematology/Oncology, Children's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
327
|
Lee KH, Lim D, Chiu S, Greenhalgh D, Cho K. Genomic landscapes of endogenous retroviruses unveil intricate genetics of conventional and genetically-engineered laboratory mouse strains. Exp Mol Pathol 2016; 100:248-56. [PMID: 26779669 DOI: 10.1016/j.yexmp.2016.01.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Accepted: 01/09/2016] [Indexed: 11/24/2022]
Abstract
Laboratory strains of mice, both conventional and genetically engineered, have been introduced as critical components of a broad range of studies investigating normal and disease biology. Currently, the genetic identity of laboratory mice is primarily confirmed by surveying polymorphisms in selected sets of "conventional" genes and/or microsatellites in the absence of a single completely sequenced mouse genome. First, we examined variations in the genomic landscapes of transposable repetitive elements, named the TREome, in conventional and genetically engineered mouse strains using murine leukemia virus-type endogenous retroviruses (MLV-ERVs) as a probe. A survey of the genomes from 56 conventional strains revealed strain-specific TREome landscapes, and certain families (e.g., C57BL) of strains were discernible with defined patterns. Interestingly, the TREome landscapes of C3H/HeJ (toll-like receptor-4 [TLR4] mutant) inbred mice were different from its control C3H/HeOuJ (TLR4 wild-type) strain. In addition, a CD14 knock-out strain had a distinct TREome landscape compared to its control/backcross C57BL/6J strain. Second, an examination of superantigen (SAg, a "TREome gene") coding sequences of mouse mammary tumor virus-type ERVs in the genomes of the 46 conventional strains revealed a high diversity, suggesting a potential role of SAgs in strain-specific immune phenotypes. The findings from this study indicate that unexplored and intricate genomic variations exist in laboratory mouse strains, both conventional and genetically engineered. The TREome-based high-resolution genetics surveillance system for laboratory mice would contribute to efficient study design with quality control and accurate data interpretation. This genetics system can be easily adapted to other species ranging from plants to humans.
Collapse
Affiliation(s)
- Kang-Hoon Lee
- Department of Surgery, University of California, Davis, and Shriners Hospitals for Children Northern California, Sacramento, CA 95817, United States
| | - Debora Lim
- Department of Surgery, University of California, Davis, and Shriners Hospitals for Children Northern California, Sacramento, CA 95817, United States
| | - Sophia Chiu
- Department of Surgery, University of California, Davis, and Shriners Hospitals for Children Northern California, Sacramento, CA 95817, United States
| | - David Greenhalgh
- Department of Surgery, University of California, Davis, and Shriners Hospitals for Children Northern California, Sacramento, CA 95817, United States
| | - Kiho Cho
- Department of Surgery, University of California, Davis, and Shriners Hospitals for Children Northern California, Sacramento, CA 95817, United States.
| |
Collapse
|
328
|
Baird A, Coimbra R, Dang X, Eliceiri BP, Costantini TW. Up-regulation of the human-specific CHRFAM7A gene in inflammatory bowel disease. BBA CLINICAL 2016; 5:66-71. [PMID: 27051591 PMCID: PMC4802402 DOI: 10.1016/j.bbacli.2015.12.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 12/16/2015] [Accepted: 12/17/2015] [Indexed: 12/16/2022]
Abstract
Background: The α7-subunit of the α7-nicotinic acetylcholine receptor (α7-nAChR) is an obligatory intermediate for the anti-inflammatory effects of the vagus nerve. But in humans, there exists a second gene called CHRFAM7A that encodes a dominant negative α7-nAChR inhibitor. Here, we investigated whether their expression was altered in inflammatory bowel disease (IBD) and colon cancer. Methods: Quantitative RT-PCR measured gene expression of human α7-nAChR gene (CHRNA7), CHRFAM7A, TBC3D1, and actin in biopsies of normal large and small intestine, and compared to their expression in biopsies of ulcerative colitis, Crohn's disease, and colon cancer. Results: qRT-PCR showed that CHRFAM7A and CHRNA7 gene expression was significantly (p < .02) up-regulated in IBD (N = 64). Gene expression was unchanged in colon cancer. Further analyses revealed that there were differences in ulcerative colitis and Crohn's Disease. Colon biopsies of ulcerative colitis (N = 33) confirmed increased expression of CHRFAM7A and decreased in CHRNA7 expression (p < 0.001). Biopsies of Crohn's disease (N = 31), however, showed only small changes in CHRFAM7A expression (p < 0.04) and no change in CHRNA7. When segregated by tissue source, both CHRFAM7A up-regulation (p < 0.02) and CHRNA7 down-regulation (p < 0.001) were measured in colon, but not in small intestine. Conclusion: The human-specific CHRFAM7A gene is up-regulated, and its target, CHRNA7, down-regulated, in IBD. Differences between ulcerative colitis and Crohn's disease tie to location of disease. Significance: The appearance of IBD in modern humans may be consequent to the emergence of CHRFAM7A, a human-specific α7-nAChR antagonist. CHRFAM7A could present a new, unrecognized target for development of IBD therapeutics. CHRFAM7A is a pro-inflammatory and human-specific gene not found in other species. CHRFAM7A expression is elevated in certain IBD, but its target CHRNA7 decreased. Changes in CHRFAM7A and CHRNA7 expression are disease- and tissue site specific. Some IBDs may be examples of “off-target disease sequelae” of human evolution. Animal modeling of human disease do not test contributions of human-specific genes.
Collapse
Affiliation(s)
- Andrew Baird
- Division of Trauma, Surgical Critical Care, Burns and Acute Care Surgery, Department of Surgery, University of California San Diego, La Jolla, CA, USA
| | - Raul Coimbra
- Division of Trauma, Surgical Critical Care, Burns and Acute Care Surgery, Department of Surgery, University of California San Diego, La Jolla, CA, USA
| | - Xitong Dang
- Division of Trauma, Surgical Critical Care, Burns and Acute Care Surgery, Department of Surgery, University of California San Diego, La Jolla, CA, USA; The Key Laboratory of Medical Electrophysiology, Institute of Cardiovascular Research, Sichuan Medical University, Luzhou, China
| | - Brian P Eliceiri
- Division of Trauma, Surgical Critical Care, Burns and Acute Care Surgery, Department of Surgery, University of California San Diego, La Jolla, CA, USA
| | - Todd W Costantini
- Division of Trauma, Surgical Critical Care, Burns and Acute Care Surgery, Department of Surgery, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
329
|
Sun J, Li N, Oh KS, Dutta B, Vayttaden SJ, Lin B, Ebert TS, De Nardo D, Davis J, Bagirzadeh R, Lounsbury NW, Pasare C, Latz E, Hornung V, Fraser IDC. Comprehensive RNAi-based screening of human and mouse TLR pathways identifies species-specific preferences in signaling protein use. Sci Signal 2016; 9:ra3. [PMID: 26732763 PMCID: PMC5381726 DOI: 10.1126/scisignal.aab2191] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Toll-like receptors (TLRs) are a major class of pattern recognition receptors, which mediate the responses of innate immune cells to microbial stimuli. To systematically determine the roles of proteins in canonical TLR signaling pathways, we conducted an RNA interference (RNAi)-based screen in human and mouse macrophages. We observed a pattern of conserved signaling module dependencies across species, but found notable species-specific requirements at the level of individual proteins. Among these, we identified unexpected differences in the involvement of members of the interleukin-1 receptor-associated kinase (IRAK) family between the human and mouse TLR pathways. Whereas TLR signaling in mouse macrophages depended primarily on IRAK4 and IRAK2, with little or no role for IRAK1, TLR signaling and proinflammatory cytokine production in human macrophages depended on IRAK1, with knockdown of IRAK4 or IRAK2 having less of an effect. Consistent with species-specific roles for these kinases, IRAK4 orthologs failed to rescue signaling in IRAK4-deficient macrophages from the other species, and only mouse macrophages required the kinase activity of IRAK4 to mediate TLR responses. The identification of a critical role for IRAK1 in TLR signaling in humans could potentially explain the association of IRAK1 with several autoimmune diseases. Furthermore, this study demonstrated how systematic screening can be used to identify important characteristics of innate immune responses across species, which could optimize therapeutic targeting to manipulate human TLR-dependent outputs.
Collapse
Affiliation(s)
- Jing Sun
- Signaling Systems Unit, Laboratory of Systems Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ning Li
- Signaling Systems Unit, Laboratory of Systems Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Kyu-Seon Oh
- Signaling Systems Unit, Laboratory of Systems Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Bhaskar Dutta
- Bioinformatics Team, Laboratory of Systems Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Sharat J Vayttaden
- Signaling Systems Unit, Laboratory of Systems Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Bin Lin
- Signaling Systems Unit, Laboratory of Systems Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Thomas S Ebert
- Institute of Molecular Medicine, University Hospital, University of Bonn, 53127 Bonn, Germany
| | - Dominic De Nardo
- Institute of Innate Immunity, University Hospital, Biomedical Centre, University of Bonn, 53127 Bonn, Germany. Inflammation Division, Walter and Eliza Hall Institute, Parkville,Victoria 3052, Australia. Department of Medical Biology, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Joie Davis
- Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Rustam Bagirzadeh
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Nicolas W Lounsbury
- Signaling Systems Unit, Laboratory of Systems Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Chandrashekhar Pasare
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Eicke Latz
- Institute of Innate Immunity, University Hospital, Biomedical Centre, University of Bonn, 53127 Bonn, Germany. German Center for Neurodegenerative Diseases (DZNE), 53175 Bonn, Germany. Department of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Veit Hornung
- Institute of Molecular Medicine, University Hospital, University of Bonn, 53127 Bonn, Germany
| | - Iain D C Fraser
- Signaling Systems Unit, Laboratory of Systems Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
330
|
Costantini TW, Meads M, Dang X, Coimbra R, Torbett BE, Baird A, Eliceiri BP. The Response to Burn Injury in Mice With Human Hematolymphoid Systems. Ann Surg 2016; 263:199-204. [PMID: 25575256 PMCID: PMC6550477 DOI: 10.1097/sla.0000000000001123] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
OBJECTIVE To develop an animal model of injury that more closely represents the human inflammatory cell response to injury. BACKGROUND Because the mouse inflammatory response to burn injury cannot account for the contribution of human-specific genes, animal models are needed to more closely recapitulate the human inflammatory response and improve the translational impact of injury research. To this end, we hypothesized that the human inflammatory cell response to injury could be selectively assessed after severe burn injury using humanized mice. METHODS NOD-Scid-IL2Rγ null mice were transplanted with human hematopoietic CD34+ progenitor cells; their engraftment confirmed and then subjected to 30% total body surface area steam burn injury. Blood, bone marrow, and lung tissue were collected 4 hours after injury and human inflammatory cell mobilization analyzed using flow cytometry and immunohistochemistry. RESULTS Burn injury caused mobilization of human inflammatory cells into the systemic circulation. Next, burn injury was accompanied by evidence of histologic lung injury and concomitant mobilization of human CD45+ immune cells into the lung that were associated with increased trafficking of human CD11b+ myeloid cells. CONCLUSIONS These experiments are the first to demonstrate the suitability of humanized mice for injury research. They offer the possibility to address very specific research questions that are not amenable to traditional mouse models of injury, for example, the emerging role of certain human-specific genes that are either unrepresented or totally absent, from the mouse genome.
Collapse
Affiliation(s)
- Todd W Costantini
- *Division of Trauma, Surgical Critical Care, Burns and Acute Care Surgery, Department of Surgery, University of California, San Diego Health Sciences, San Diego, CA †Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA
| | | | | | | | | | | | | |
Collapse
|
331
|
Stolfi JL, Pai CCS, Murphy WJ. Preclinical modeling of hematopoietic stem cell transplantation - advantages and limitations. FEBS J 2015; 283:1595-606. [PMID: 26640088 DOI: 10.1111/febs.13612] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Revised: 11/13/2015] [Accepted: 11/25/2015] [Indexed: 12/31/2022]
Abstract
Hematopoietic stem cell transplantation, which was first successfully performed in the 1950s, remains a critical therapeutic modality for treatment of a diverse array of diseases, including a multitude of hematological malignancies, autoimmune disorders, amyloidosis and inherited genetic hematological disorders. Although great advances have been made in understanding and application of this therapy, significant complications still exist, warranting further investigation. Of critical importance, graft-versus-host disease (GVHD), in both acute and chronic forms, remains a major complication of hematopoietic stem cell transplantation, responsible for both the development of chronic illness and morbidity, as well as mortality. Use of an appropriate preclinical model may provide significant insight into the mechanistic pathways leading to the development and progression of graft-versus-host disease, as well as cancer in general. However, existing preclinical modeling systems exhibit significant limitations, and development of models that recapitulate the complex and comprehensive clinical scenario and provide a tool by which therapeutic intervention may be developed and assessed is of utmost importance. Here, we review the present status of the field of graft-versus-host disease research. We discuss and summarize the preclinical models currently in use, as well as their advantages and limitations.
Collapse
Affiliation(s)
- Jessica L Stolfi
- Department of Dermatology, School of Medicine, University of California, Davis, Sacramento, CA, USA
| | - Chien-Chun S Pai
- Department of Dermatology, School of Medicine, University of California, Davis, Sacramento, CA, USA
| | - William J Murphy
- Department of Dermatology, School of Medicine, University of California, Davis, Sacramento, CA, USA.,Department of Internal Medicine, School of Medicine, University of California, Davis, Sacramento, CA, USA
| |
Collapse
|
332
|
Plasma Glycoproteomics Reveals Sepsis Outcomes Linked to Distinct Proteins in Common Pathways. Crit Care Med 2015; 43:2049-2058. [PMID: 26086942 DOI: 10.1097/ccm.0000000000001134] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
OBJECTIVE Sepsis remains a predominant cause of mortality in the ICU, yet strategies to increase survival have proved largely unsuccessful. This study aimed to identify proteins linked to sepsis outcomes using a glycoproteomic approach to target extracellular proteins that trigger downstream pathways and direct patient outcomes. DESIGN Plasma was obtained from the Lactate Assessment in the Treatment of Early Sepsis cohort. N-linked plasma glycopeptides were quantified by solid-phase extraction coupled with mass spectrometry. Glycopeptides were assigned to proteins using RefSeq (National Center of Biotechnology Information, Bethesda, MD) and visualized in a heat map. Protein differences were validated by immunoblotting, and proteins were mapped for biological processes using Database for Annotation, Visualization and Integrated Discovery (National Institute of Allergy and Infectious Diseases, National Institutes of Health; Bethesda, MD) and for functional pathways using Kyoto Encyclopedia of Genes and Genomes (Kanehisa Laboratories, Kyoto, Japan) databases. SETTING Hospitalized care. PATIENTS Patients admitted to the emergency department were enrolled in the study when the diagnosis of sepsis was made, within 6 hours of presentation. INTERVENTIONS None. MEASUREMENTS AND MAIN RESULTS A total of 501 glycopeptides corresponding to 234 proteins were identified. Of these, 66 glycopeptides were unique to the survivor group and corresponded to 54 proteins, 60 were unique to the nonsurvivor group and corresponded to 43 proteins, and 375 were common responses between groups and corresponded to 137 proteins. Immunoblotting showed that nonsurvivors had increased total kininogen; decreased total cathepsin-L1, vascular cell adhesion molecule, periostin, and neutrophil gelatinase-associated lipocalin; and a two-fold decrease in glycosylated clusterin (all p < 0.05). Kyoto Encyclopedia of Genes and Genomes analysis identified six enriched pathways. Interestingly, survivors relied on the extrinsic pathway of the complement and coagulation cascade, whereas nonsurvivors relied on the intrinsic pathway. CONCLUSION This study identifies proteins linked to patient outcomes and provides insight into unexplored mechanisms that can be investigated for the identification of novel therapeutic targets.
Collapse
|
333
|
Immune regulation of kidney disease in 2015: Updates on immunosuppression in kidney disease. Nat Rev Nephrol 2015; 12:65-6. [PMID: 26656459 DOI: 10.1038/nrneph.2015.202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
334
|
Namas RA, Mi Q, Namas R, Almahmoud K, Zaaqoq AM, Abdul-Malak O, Azhar N, Day J, Abboud A, Zamora R, Billiar TR, Vodovotz Y. Insights into the Role of Chemokines, Damage-Associated Molecular Patterns, and Lymphocyte-Derived Mediators from Computational Models of Trauma-Induced Inflammation. Antioxid Redox Signal 2015; 23:1370-87. [PMID: 26560096 PMCID: PMC4685502 DOI: 10.1089/ars.2015.6398] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
SIGNIFICANCE Traumatic injury elicits a complex, dynamic, multidimensional inflammatory response that is intertwined with complications such as multiple organ dysfunction and nosocomial infection. The complex interplay between inflammation and physiology in critical illness remains a challenge for translational research, including the extrapolation to human disease from animal models. RECENT ADVANCES Over the past decade, we and others have attempted to decipher the biocomplexity of inflammation in these settings of acute illness, using computational models to improve clinical translation. In silico modeling has been suggested as a computationally based framework for integrating data derived from basic biology experiments as well as preclinical and clinical studies. CRITICAL ISSUES Extensive studies in cells, mice, and human blunt trauma patients have led us to suggest (i) that while an adequate level of inflammation is required for healing post-trauma, inflammation can be harmful when it becomes self-sustaining via a damage-associated molecular pattern/Toll-like receptor-driven feed-forward circuit; (ii) that chemokines play a central regulatory role in driving either self-resolving or self-maintaining inflammation that drives the early activation of both classical innate and more recently recognized lymphoid pathways; and (iii) the presence of multiple thresholds and feedback loops, which could significantly affect the propagation of inflammation across multiple body compartments. FUTURE DIRECTIONS These insights from data-driven models into the primary drivers and interconnected networks of inflammation have been used to generate mechanistic computational models. Together, these models may be used to gain basic insights as well as serving to help define novel biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Rami A. Namas
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
- Center for Inflammation and Regenerative Modeling, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Qi Mi
- Department of Sports Medicine and Nutrition, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Rajaie Namas
- Department of Internal Medicine, Division of Rheumatology, University of Michigan, Ann Arbor, Michigan
| | - Khalid Almahmoud
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Akram M. Zaaqoq
- Department of Critical Care Medicine, University of Pittsburgh, Pennsylvania
| | - Othman Abdul-Malak
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Nabil Azhar
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Judy Day
- Department of Mathematics, University of Tennessee, Knoxville, Tennessee
| | - Andrew Abboud
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Ruben Zamora
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Timothy R. Billiar
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
- Center for Inflammation and Regenerative Modeling, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Yoram Vodovotz
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
- Center for Inflammation and Regenerative Modeling, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
335
|
Mellanby RJ. Our time is now - how companion animal veterinarians can transform biomedical science. J Small Anim Pract 2015; 56:689-92. [DOI: 10.1111/jsap.12421] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Accepted: 10/21/2015] [Indexed: 12/19/2022]
Affiliation(s)
- R. J. Mellanby
- Division of Veterinary Clinical Studies, Easter Bush Veterinary Centre, Royal (Dick) School of Veterinary Studies and The Roslin Institute; The University of Edinburgh, Hospital for Small Animals; Roslin Midlothian EH25 9RG
| |
Collapse
|
336
|
Szczesny B, Brunyánszki A, Ahmad A, Oláh G, Porter C, Toliver-Kinsky T, Sidossis L, Herndon DN, Szabo C. Time-Dependent and Organ-Specific Changes in Mitochondrial Function, Mitochondrial DNA Integrity, Oxidative Stress and Mononuclear Cell Infiltration in a Mouse Model of Burn Injury. PLoS One 2015; 10:e0143730. [PMID: 26630679 PMCID: PMC4668069 DOI: 10.1371/journal.pone.0143730] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Accepted: 11/08/2015] [Indexed: 01/11/2023] Open
Abstract
Severe thermal injury induces a pathophysiological response that affects most of the organs within the body; liver, heart, lung, skeletal muscle among others, with inflammation and hyper-metabolism as a hallmark of the post-burn damage. Oxidative stress has been implicated as a key component in development of inflammatory and metabolic responses induced by burn. The goal of the current study was to evaluate several critical mitochondrial functions in a mouse model of severe burn injury. Mitochondrial bioenergetics, measured by Extracellular Flux Analyzer, showed a time dependent, post-burn decrease in basal respiration and ATP-turnover but enhanced maximal respiratory capacity in mitochondria isolated from the liver and lung of animals subjected to burn injury. Moreover, we detected a tissue-specific degree of DNA damage, particularly of the mitochondrial DNA, with the most profound effect detected in lungs and hearts of mice subjected to burn injury. Increased mitochondrial biogenesis in lung tissue in response to burn injury was also observed. Burn injury also induced time dependent increases in oxidative stress (measured by amount of malondialdehyde) and neutrophil infiltration (measured by myeloperoxidase activity), particularly in lung and heart. Tissue mononuclear cell infiltration was also confirmed by immunohistochemistry. The amount of poly(ADP-ribose) polymers decreased in the liver, but increased in the heart in later time points after burn. All of these biochemical changes were also associated with histological alterations in all three organs studied. Finally, we detected a significant increase in mitochondrial DNA fragments circulating in the blood immediately post-burn. There was no evidence of systemic bacteremia, or the presence of bacterial DNA fragments at any time after burn injury. The majority of the measured parameters demonstrated a sustained elevation even at 20–40 days post injury suggesting a long-lasting effect of thermal injury on organ function. The current data show that there are marked time-dependent and tissue-specific alterations in mitochondrial function induced by thermal injury, and suggest that mitochondria-specific damage is one of the earliest responses to burn injury. Mitochondria may be potential therapeutic targets in the future experimental therapy of burns.
Collapse
Affiliation(s)
- Bartosz Szczesny
- Department of Anesthesiology, The University of Texas Medical Branch, Galveston, TX, United States of America.,Shriners Hospitals for Children, Galveston, TX, United States of America
| | - Attila Brunyánszki
- Department of Anesthesiology, The University of Texas Medical Branch, Galveston, TX, United States of America
| | - Akbar Ahmad
- Department of Anesthesiology, The University of Texas Medical Branch, Galveston, TX, United States of America
| | - Gabor Oláh
- Department of Anesthesiology, The University of Texas Medical Branch, Galveston, TX, United States of America
| | - Craig Porter
- Shriners Hospitals for Children, Galveston, TX, United States of America.,Department of Surgery, The University of Texas Medical Branch, Galveston, TX, United States of America
| | - Tracy Toliver-Kinsky
- Department of Anesthesiology, The University of Texas Medical Branch, Galveston, TX, United States of America.,Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, TX, United States of America
| | - Labros Sidossis
- Shriners Hospitals for Children, Galveston, TX, United States of America.,Department of Surgery, The University of Texas Medical Branch, Galveston, TX, United States of America
| | - David N Herndon
- Shriners Hospitals for Children, Galveston, TX, United States of America.,Department of Surgery, The University of Texas Medical Branch, Galveston, TX, United States of America
| | - Csaba Szabo
- Department of Anesthesiology, The University of Texas Medical Branch, Galveston, TX, United States of America.,Shriners Hospitals for Children, Galveston, TX, United States of America
| |
Collapse
|
337
|
Efron PA, Mohr AM, Moore FA, Moldawer LL. The future of murine sepsis and trauma research models. J Leukoc Biol 2015; 98:945-52. [PMID: 26034205 PMCID: PMC4661039 DOI: 10.1189/jlb.5mr0315-127r] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Revised: 04/24/2015] [Accepted: 05/06/2015] [Indexed: 12/23/2022] Open
Abstract
Recent comparisons of the murine and human transcriptome in health and disease have called into question the appropriateness of the use of murine models for human sepsis and trauma research. More specifically, researchers have debated the suitability of mouse models of severe inflammation that is intended for eventual translation to human patients. This mini-review outlines this recent research, as well as specifically defines the arguments for and against murine models of sepsis and trauma research based on these transcriptional studies. In addition, we review newer advancements in murine models of infection and injury and define what we envision as an evolving but viable future for murine studies of sepsis and trauma.
Collapse
Affiliation(s)
- Philip A Efron
- Department of Surgery, University of Florida College of Medicine, Gainesville, Florida, USA
| | - Alicia M Mohr
- Department of Surgery, University of Florida College of Medicine, Gainesville, Florida, USA
| | - Frederick A Moore
- Department of Surgery, University of Florida College of Medicine, Gainesville, Florida, USA
| | - Lyle L Moldawer
- Department of Surgery, University of Florida College of Medicine, Gainesville, Florida, USA
| |
Collapse
|
338
|
Robert C, Kapetanovic R, Beraldi D, Watson M, Archibald AL, Hume DA. Identification and annotation of conserved promoters and macrophage-expressed genes in the pig genome. BMC Genomics 2015; 16:970. [PMID: 26582032 PMCID: PMC4652390 DOI: 10.1186/s12864-015-2111-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Accepted: 10/19/2015] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND The FANTOM5 consortium used Cap Analysis of Gene Expression (CAGE) tag sequencing to produce a comprehensive atlas of promoters and enhancers within the human and mouse genomes. We reasoned that the mapping of these regulatory elements to the pig genome could provide useful annotation and evidence to support assignment of orthology. RESULTS For human transcription start sites (TSS) associated with annotated human-mouse orthologs, 17% mapped to the pig genome but not to the mouse, 10% mapped only to the mouse, and 55% mapped to both pig and mouse. Around 17% did not map to either species. The mapping percentages were lower where there was not clear orthology relationship, but in every case, mapping to pig was greater than to mouse, and the degree of homology was also greater. Combined mapping of mouse and human CAGE-defined promoters identified at least one putative conserved TSS for >16,000 protein-coding genes. About 54% of the predicted locations of regulatory elements in the pig genome were supported by CAGE and/or RNA-Seq analysis from pig macrophages. CONCLUSIONS Comparative mapping of promoters and enhancers from humans and mice can provide useful preliminary annotation of other animal genomes. The data also confirm extensive gain and loss of regulatory elements between species, and the likelihood that pigs provide a better model than mice for human gene regulation and function.
Collapse
Affiliation(s)
- Christelle Robert
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, EH25 9RG, Edinburgh, UK.
| | - Ronan Kapetanovic
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia.
| | - Dario Beraldi
- Cancer Research UK, Cambridge Research Institute, Li Ka Shing Center, Robinson Way, Cambridge, CB2 0RE, UK.
| | - Mick Watson
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, EH25 9RG, Edinburgh, UK.
- Edinburgh Genomics, University of Edinburgh, Easter Bush, Edinburgh, EH25 9RG, UK.
| | - Alan L Archibald
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, EH25 9RG, Edinburgh, UK.
| | - David A Hume
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, EH25 9RG, Edinburgh, UK.
| |
Collapse
|
339
|
Mira JC, Szpila BE, Nacionales DC, Lopez MC, Gentile LF, Mathias BJ, Vanzant EL, Ungaro R, Holden D, Rosenthal MD, Rincon J, Verdugo PT, Larson SD, Moore FA, Brakenridge SC, Mohr AM, Baker HV, Moldawer LL, Efron PA. Patterns of gene expression among murine models of hemorrhagic shock/trauma and sepsis. Physiol Genomics 2015; 48:135-44. [PMID: 26578697 DOI: 10.1152/physiolgenomics.00072.2015] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 11/13/2015] [Indexed: 01/22/2023] Open
Abstract
Controversy remains whether the leukocyte genomic response to trauma or sepsis is dependent upon the initiating stimulus. Previous work illustrated poor correlations between historical models of murine trauma and sepsis (i.e., trauma-hemorrhage and lipopolysaccharide injection, respectively). The aim of this study is to examine the early genomic response in improved murine models of sepsis [cecal ligation and puncture (CLP)] and trauma [polytrauma (PT)] with and without pneumonia (PT+Pp). Groups of naïve, CLP, PT, and PT+Pp mice were killed at 2 h, 1 or 3 days. Total leukocytes were isolated for genome-wide expression analysis, and genes that were found to differ from control (false discovery rate adjusted P < 0.001) were assessed for fold-change differences. Spearman correlations were also performed. For all time points combined (CLP, PT, PT+Pp), there were 10,426 total genes that were found to significantly differ from naïve controls. At 2 h, the transcriptomic changes between CLP and PT showed a positive correlation (rs) of 0.446 (P < 0.0001) but were less positive thereafter. Correlations were significantly improved when we limited the analysis to common genes whose expression differed by a 1.5 fold-change. Both pathway and upstream analyses revealed the activation of genes known to be associated with pathogen-associated and damage-associated molecular pattern signaling, and early activation patterns of expression were very similar between polytrauma and sepsis at the earliest time points. This study demonstrates that the early leukocyte genomic response to sepsis and trauma are very similar in mice.
Collapse
Affiliation(s)
- Juan C Mira
- Department of Surgery, University of Florida College of Medicine, Gainesville, Florida; and
| | - Benjamin E Szpila
- Department of Surgery, University of Florida College of Medicine, Gainesville, Florida; and
| | - Dina C Nacionales
- Department of Surgery, University of Florida College of Medicine, Gainesville, Florida; and
| | - Maria-Cecilia Lopez
- Department of Molecular Genetics and Microbiology, University of Florida College of Medicine, Gainesville, Florida
| | - Lori F Gentile
- Department of Surgery, University of Florida College of Medicine, Gainesville, Florida; and
| | - Brittany J Mathias
- Department of Surgery, University of Florida College of Medicine, Gainesville, Florida; and
| | - Erin L Vanzant
- Department of Surgery, University of Florida College of Medicine, Gainesville, Florida; and
| | - Ricardo Ungaro
- Department of Surgery, University of Florida College of Medicine, Gainesville, Florida; and
| | - David Holden
- Department of Surgery, University of Florida College of Medicine, Gainesville, Florida; and
| | - Martin D Rosenthal
- Department of Surgery, University of Florida College of Medicine, Gainesville, Florida; and
| | - Jaimar Rincon
- Department of Surgery, University of Florida College of Medicine, Gainesville, Florida; and
| | - Patrick T Verdugo
- Department of Surgery, University of Florida College of Medicine, Gainesville, Florida; and
| | - Shawn D Larson
- Department of Surgery, University of Florida College of Medicine, Gainesville, Florida; and
| | - Frederick A Moore
- Department of Surgery, University of Florida College of Medicine, Gainesville, Florida; and
| | - Scott C Brakenridge
- Department of Surgery, University of Florida College of Medicine, Gainesville, Florida; and
| | - Alicia M Mohr
- Department of Surgery, University of Florida College of Medicine, Gainesville, Florida; and
| | - Henry V Baker
- Department of Molecular Genetics and Microbiology, University of Florida College of Medicine, Gainesville, Florida
| | - Lyle L Moldawer
- Department of Surgery, University of Florida College of Medicine, Gainesville, Florida; and
| | - Philip A Efron
- Department of Surgery, University of Florida College of Medicine, Gainesville, Florida; and
| |
Collapse
|
340
|
Akscyn RM, Franklin JL, Gavrikova TA, Schwacha MG, Messina JL. A rat model of concurrent combined injuries (polytrauma). Int J Clin Exp Med 2015; 8:20097-20110. [PMID: 26884923 PMCID: PMC4723768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 11/05/2015] [Indexed: 06/05/2023]
Abstract
Polytrauma, a combination of injuries to more than one body part or organ system, is common in modern warfare and in automobile and industrial accidents. The combination of injuries can include burn injury, fracture, hemorrhage, trauma to the extremities, and trauma to specific organ systems. To investigate the effects of combined injuries, we have developed a new and highly reproducible model of polytrauma. This model combines burn injury with soft tissue and gastrointestinal (GI) tract trauma. Male Sprague Dawley rats were subjected to a 15-20% total body surface area scald burn, or a single puncture of the cecum with a G30 needle, or the combination of both injuries (polytrauma). Unlike many 'double hit' models, the injuries in our model were performed simultaneously. We asked whether multiple minor injuries, when combined, would result in a distinct phenotype, different from single minor injuries or a more severe single injury. There were differences between the single injuries and polytrauma in the maintenance of blood glucose, body temperature, body weight, hepatic mRNA and circulating levels of TNF-α, IL-1β and IL-6, and hepatic ER-stress. It has been suggested that models utilizing combinatorial injuries may be needed to more accurately model the human condition. We believe our model is ideal for studying the complex sequelae of polytrauma, which differs from single injuries. Insights gained from this model may suggest better treatment options to improve patient outcomes.
Collapse
Affiliation(s)
- Robert M Akscyn
- Department of Pathology, Division of Molecular and Cellular Pathology, University of Alabama at BirminghamBirmingham, Alabama, 35294-0019
| | - J Lee Franklin
- Department of Pathology, Division of Molecular and Cellular Pathology, University of Alabama at BirminghamBirmingham, Alabama, 35294-0019
| | - Tatyana A Gavrikova
- Department of Pathology, Division of Molecular and Cellular Pathology, University of Alabama at BirminghamBirmingham, Alabama, 35294-0019
| | - Martin G Schwacha
- Department of Surgery, Division of Trauma and Emergency Surgery, University of Texas Health Science Center San AntonioSan Antonio, Texas, 78229-3900
- US Army Institute of Surgical ResearchFort Sam Houston, Texas, 78234
| | - Joseph L Messina
- Department of Pathology, Division of Molecular and Cellular Pathology, University of Alabama at BirminghamBirmingham, Alabama, 35294-0019
- Veterans Affairs Medical CenterBirmingham, AL 35233
| |
Collapse
|
341
|
Wessels I, Cousins RJ. Zinc dyshomeostasis during polymicrobial sepsis in mice involves zinc transporter Zip14 and can be overcome by zinc supplementation. Am J Physiol Gastrointest Liver Physiol 2015; 309:G768-78. [PMID: 26272258 PMCID: PMC4628964 DOI: 10.1152/ajpgi.00179.2015] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Accepted: 07/15/2015] [Indexed: 01/31/2023]
Abstract
Integrity of the immune system is particularly dependent on the availability of zinc. Recent data suggest that zinc is involved in the development of sepsis, a life-threatening systemic inflammation with high death rates, but with limited therapeutic options. Altered cell zinc transport mechanisms could contribute to the inflammatory effects of sepsis. Zip14, a zinc importer induced by proinflammatory stimuli, could influence zinc metabolism during sepsis and serve as a target for therapy. Using cecal ligation-and-puncture (CLP) to model polymicrobial sepsis, we narrowed the function of ZIP14 to regulation of zinc homeostasis in hepatocytes, while hepatic leukocytes were mostly responsible for driving inflammation, as shown by higher expression of IL-1β, TNFα, S100A8, and matrix metalloproteinase-8. Using Zip14 knockout (KO) mice as a novel approach, we found that ablation of Zip14 produced a delay in development of leukocytosis, prevented zinc accumulation in the liver, altered the kinetics of hypozincemia, and drastically increased serum IL-6, TNFα, and IL-10 concentrations following CLP. Hence, this model revealed that the zinc transporter ZIP14 is a component of the pathway for zinc redistribution that contributes to zinc dyshomeostasis during polymicrobial sepsis. In contrast, using the identical CLP model, we found that supplemental dietary zinc reduced the severity of sepsis, as shown by amelioration of cytokines, calprotectins, and blood bacterial loads. We conclude that the zinc transporter ZIP14 influences aspects of the pathophysiology of nonlethal polymicrobial murine sepsis induced by CLP through zinc delivery. The results are promising for the use of zinc and its transporters as targets for future sepsis therapy.
Collapse
Affiliation(s)
- Inga Wessels
- Food Science and Human Nutrition Department, Center for Nutritional Sciences, College of Agricultural and Life Sciences, University of Florida, Gainesville, Florida
| | - Robert J. Cousins
- Food Science and Human Nutrition Department, Center for Nutritional Sciences, College of Agricultural and Life Sciences, University of Florida, Gainesville, Florida
| |
Collapse
|
342
|
Zou L, Chen HH, Li D, Xu G, Feng Y, Chen C, Wang L, Sosnovik DE, Chao W. Imaging Lymphoid Cell Death In Vivo During Polymicrobial Sepsis. Crit Care Med 2015; 43:2303-12. [PMID: 26335111 PMCID: PMC6889962 DOI: 10.1097/ccm.0000000000001254] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
OBJECTIVES Cell death in lymphatic organs, such as the spleen, is in part responsible for immunosuppression and contributes to mortality during sepsis. An early and noninvasive detection of lymphoid cell death could thus have significant clinical implications. Here, we tested in vivo imaging of lymphoid cell death using a near-infrared annexin V (AV-750). DESIGN Animal study. SETTING Laboratory investigation. SUBJECTS C57BL/6J wild-type and toll-like receptor 3 knockout mice. INTERVENTIONS Mild and severe polymicrobial sepsis was induced with cecum ligation and puncture. Serum cytokines and acute kidney injury markers were tested by immunoassay and quantitative reverse transcription-polymerase chain reaction, respectively. Sepsis-induced lymphoid cell death was detected by fluorescent AV-750 accumulation in the thorax and abdomen (in vivo), in isolated organs (ex vivo), and in isolated cells (flow cytometry). Caspase-3 cleavage/activity and terminal deoxynucleotidyl transferase dUTP nick-end labeling staining were tested for apoptosis. MEASUREMENTS AND MAIN RESULTS Severe sepsis induced marked apoptosis in the thymus, spleen, and liver as demonstrated by cleaved caspase-3 and an increase in caspase-3 activity and terminal deoxynucleotidyl transferase dUTP nick-end labeling-positive cells. A significant increase in fluorescent AV-750 signal was seen in the thoracic and upper abdominal fields and associated with the severity of sepsis. The in vivo thoracic and abdominal AV-750 fluorescent signal was attributed to the thymus, liver, and spleen as determined by ex vivo imaging and highly correlated with the levels of cell death in thymocytes and splenocytes, respectively, as measured by flow cytometry. Compared with wild-type septic mice, toll-like receptor 3 septic mice had attenuated abdominal AV-750 fluorescent signal, reduced ex vivo fluorescence in the spleen, and decreased splenocyte cell death. CONCLUSIONS In vivo AV-750 fluorescent imaging provides spatially resolved and organ-specific detection of lymphoid cell death during polymicrobial sepsis. The AV-750 fluorescent intensity in the thoracic and abdominal fields is associated with sepsis severity and well correlated with sepsis-induced cell death in the thymus and spleen, respectively.
Collapse
Affiliation(s)
- Lin Zou
- 1Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA. 2Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA. 3Department of Pathology and Laboratory Medicine, Children's Hospital Los Angeles, Los Angeles, CA
| | | | | | | | | | | | | | | | | |
Collapse
|
343
|
Hazeldine J, Lord JM, Hampson P. Immunesenescence and inflammaging: A contributory factor in the poor outcome of the geriatric trauma patient. Ageing Res Rev 2015; 24:349-57. [PMID: 26484895 DOI: 10.1016/j.arr.2015.10.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Revised: 10/12/2015] [Accepted: 10/13/2015] [Indexed: 12/13/2022]
Abstract
Compared to younger patients, traumatic injury in older patients is associated with increased mortality and a range of adverse outcomes such as higher rates of infectious episodes, longer length of hospital stay and poor functional outcome at follow up. Data emerging from human and murine-based studies suggest age-related changes in immune function, collectively termed immunesenescence, and the chronic sub-clinical systemic inflammatory state of older adults, termed inflammaging, may contribute to these poor outcomes. Here, we review the findings of these studies, whose results demonstrate that the geriatric trauma patient elicits an immune response to injury that is distinct to that of younger adults, being characterised by reduced immune cell activation, impaired function and abnormal haematopoiesis, defects that are accompanied by an altered inflammatory response that fails to return to a homeostatic baseline in the days following injury. Although considerable evidence is accumulating that demonstrates clear and significant age-related differences in the immune and inflammatory response to traumatic injury, our current understanding of the mechanism(s) that underlie these changes is limited. Future studies that provide a mechanistic explanation for the unique immune and inflammatory response of older adults to traumatic injury are therefore essential if we are to determine whether manipulation of the immune system has potential as a future therapeutic strategy by which to improve the outcome of the geriatric trauma patient.
Collapse
|
344
|
Spinelli L, Carpentier S, Montañana Sanchis F, Dalod M, Vu Manh TP. BubbleGUM: automatic extraction of phenotype molecular signatures and comprehensive visualization of multiple Gene Set Enrichment Analyses. BMC Genomics 2015; 16:814. [PMID: 26481321 PMCID: PMC4617899 DOI: 10.1186/s12864-015-2012-4] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Accepted: 10/07/2015] [Indexed: 12/21/2022] Open
Abstract
Background Recent advances in the analysis of high-throughput expression data have led to the development of tools that scaled-up their focus from single-gene to gene set level. For example, the popular Gene Set Enrichment Analysis (GSEA) algorithm can detect moderate but coordinated expression changes of groups of presumably related genes between pairs of experimental conditions. This considerably improves extraction of information from high-throughput gene expression data. However, although many gene sets covering a large panel of biological fields are available in public databases, the ability to generate home-made gene sets relevant to one’s biological question is crucial but remains a substantial challenge to most biologists lacking statistic or bioinformatic expertise. This is all the more the case when attempting to define a gene set specific of one condition compared to many other ones. Thus, there is a crucial need for an easy-to-use software for generation of relevant home-made gene sets from complex datasets, their use in GSEA, and the correction of the results when applied to multiple comparisons of many experimental conditions. Result We developed BubbleGUM (GSEA Unlimited Map), a tool that allows to automatically extract molecular signatures from transcriptomic data and perform exhaustive GSEA with multiple testing correction. One original feature of BubbleGUM notably resides in its capacity to integrate and compare numerous GSEA results into an easy-to-grasp graphical representation. We applied our method to generate transcriptomic fingerprints for murine cell types and to assess their enrichments in human cell types. This analysis allowed us to confirm homologies between mouse and human immunocytes. Conclusions BubbleGUM is an open-source software that allows to automatically generate molecular signatures out of complex expression datasets and to assess directly their enrichment by GSEA on independent datasets. Enrichments are displayed in a graphical output that helps interpreting the results. This innovative methodology has recently been used to answer important questions in functional genomics, such as the degree of similarities between microarray datasets from different laboratories or with different experimental models or clinical cohorts. BubbleGUM is executable through an intuitive interface so that both bioinformaticians and biologists can use it. It is available at http://www.ciml.univ-mrs.fr/applications/BubbleGUM/index.html. Electronic supplementary material The online version of this article (doi:10.1186/s12864-015-2012-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Lionel Spinelli
- Centre d'Immunologie, de Marseille-Luminy, Aix Marseille University UM2, Inserm, U1104, CNRS UMR7280, F-13288, Marseille, Cedex 09, France.
| | | | - Frédéric Montañana Sanchis
- Centre d'Immunologie, de Marseille-Luminy, Aix Marseille University UM2, Inserm, U1104, CNRS UMR7280, F-13288, Marseille, Cedex 09, France.
| | - Marc Dalod
- Centre d'Immunologie, de Marseille-Luminy, Aix Marseille University UM2, Inserm, U1104, CNRS UMR7280, F-13288, Marseille, Cedex 09, France.
| | - Thien-Phong Vu Manh
- Centre d'Immunologie, de Marseille-Luminy, Aix Marseille University UM2, Inserm, U1104, CNRS UMR7280, F-13288, Marseille, Cedex 09, France.
| |
Collapse
|
345
|
Leelahavanichkul A, Somparn P, Bootprapan T, Tu H, Tangtanatakul P, Nuengjumnong R, Worasilchai N, Tiranathanagul K, Eiam-ong S, Levine M, Chinampon A, Srisawat N. High-dose ascorbate with low-dose amphotericin B attenuates severity of disease in a model of the reappearance of candidemia during sepsis in the mouse. Am J Physiol Regul Integr Comp Physiol 2015; 309:R223-34. [PMID: 25994956 DOI: 10.1152/ajpregu.00238.2014] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Amphotericin B (Ampho B) isa fungicidal drug that causes cell wall injury. Pharmacological ascorbate induces the extracellular prooxidants, which might enter the Ampho B-induced cell wall porosity and act synergistically.W e tested low-dose Ampho B with a short course of pharmacological ascorbate using a mouse model of sepsis preconditioned with an injection of Candida albicans 6 h prior to cecal ligation and puncture (CLP). In this model, candidemia reappeared as early as 6 h after CLP with a predictably high mortality rate. This characteristic mimics sepsis in the phase of immunosuppression inpatients. Using the model, at 12- and 18-h post-CLP, we administered isotonic (pH neutralized) pharmacological ascorbate intravenously with low-dose Ampho B or sodium deoxycholate, vehicle-controlled, administered IP. The survival rate of low-dose Ampho B plus ascorbate was 53%, compared with < 11% for low-dose Ampho B or high-dose Ampho B alone. In addition, a beneficial effect was demonstrated in terms of kidney damage,liver injury, spleen histopathology, and serum markers at 24 h after CLP. Kidney injury was less severe in low-dose Ampho B plus ascorbate combination therapy due to less severe sepsis. Moreover, ascorbate enhanced the effectiveness of phagocytosis against C. albicans in human phagocytic cells. Taken together, the data indicate that the new mouse model simulates sepsis-induced immunosuppression and that the combination of pharmacological ascorbate with an antifungal drug is a potentially effective treatment that may reduce nephrotoxicity, and perhaps also increase fungicidal activity in patients with systemic candidiasis caused by Candida albicans.
Collapse
|
346
|
Malkin AD, Sheehan RP, Mathew S, Federspiel WJ, Redl H, Clermont G. A Neutrophil Phenotype Model for Extracorporeal Treatment of Sepsis. PLoS Comput Biol 2015; 11:e1004314. [PMID: 26468651 PMCID: PMC4607502 DOI: 10.1371/journal.pcbi.1004314] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Accepted: 05/01/2015] [Indexed: 11/18/2022] Open
Abstract
Neutrophils play a central role in eliminating bacterial pathogens, but may also contribute to end-organ damage in sepsis. Interleukin-8 (IL-8), a key modulator of neutrophil function, signals through neutrophil specific surface receptors CXCR-1 and CXCR-2. In this study a mechanistic computational model was used to evaluate and deploy an extracorporeal sepsis treatment which modulates CXCR-1/2 levels. First, a simplified mechanistic computational model of IL-8 mediated activation of CXCR-1/2 receptors was developed, containing 16 ODEs and 43 parameters. Receptor level dynamics and systemic parameters were coupled with multiple neutrophil phenotypes to generate dynamic populations of activated neutrophils which reduce pathogen load, and/or primed neutrophils which cause adverse tissue damage when misdirected. The mathematical model was calibrated using experimental data from baboons administered a two-hour infusion of E coli and followed for a maximum of 28 days. Ensembles of parameters were generated using a Bayesian parallel tempering approach to produce model fits that could recreate experimental outcomes. Stepwise logistic regression identified seven model parameters as key determinants of mortality. Sensitivity analysis showed that parameters controlling the level of killer cell neutrophils affected the overall systemic damage of individuals. To evaluate rescue strategies and provide probabilistic predictions of their impact on mortality, time of onset, duration, and capture efficacy of an extracorporeal device that modulated neutrophil phenotype were explored. Our findings suggest that interventions aiming to modulate phenotypic composition are time sensitive. When introduced between 3–6 hours of infection for a 72 hour duration, the survivor population increased from 31% to 40–80%. Treatment efficacy quickly diminishes if not introduced within 15 hours of infection. Significant harm is possible with treatment durations ranging from 5–24 hours, which may reduce survival to 13%. In severe sepsis, an extracorporeal treatment which modulates CXCR-1/2 levels has therapeutic potential, but also potential for harm. Further development of the computational model will help guide optimal device development and determine which patient populations should be targeted by treatment. Sepsis occurs when a patient develops a whole body immune response due to infection. In this condition, white blood cells called neutrophils circulate in an active state, seeking and eliminating invading bacteria. However, when neutrophils are activated, healthy tissue is inadvertently targeted, leading to organ damage and potentially death. Even though sepsis kills millions worldwide, there are still no specific treatments approved in the United States. This may be due to the complexity and diversity of the body’s immune response, which can be managed well using computational modeling. We have developed a computational model to predict how different levels of neutrophil activation impact survival in an overactive inflammatory conditions. The model was utilized to assess the effectiveness of a simulated experimental sepsis treatment which modulates neutrophil populations and activity. This evaluation determined that treatment timing plays a critical role in therapeutic effectiveness. When utilized properly the treatment drastically improves survival, but there is also risk of causing patient harm when introduced at the wrong time. We intend for this computational model to support and guide further development of sepsis treatments and help translate these preliminary results from bench to bedside.
Collapse
Affiliation(s)
- Alexander D. Malkin
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- * E-mail:
| | - Robert P. Sheehan
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Shibin Mathew
- Department of Chemical and Petroleum Engineering, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - William J. Federspiel
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Chemical and Petroleum Engineering, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Heinz Redl
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology in AUVA center, Vienna, Austria
| | - Gilles Clermont
- CRISMA Center, Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| |
Collapse
|
347
|
Hawiger J, Veach RA, Zienkiewicz J. New paradigms in sepsis: from prevention to protection of failing microcirculation. J Thromb Haemost 2015; 13:1743-56. [PMID: 26190521 PMCID: PMC5014149 DOI: 10.1111/jth.13061] [Citation(s) in RCA: 95] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Accepted: 07/06/2015] [Indexed: 12/24/2022]
Abstract
Sepsis, also known as septicemia, is one of the 10 leading causes of death worldwide. The rising tide of sepsis due to bacterial, fungal and viral infections cannot be stemmed by current antimicrobial therapies and supportive measures. New paradigms for the mechanism and resolution of sepsis and consequences for sepsis survivors are emerging. Consistent with Benjamin Franklin's dictum 'an ounce of prevention is worth a pound of cure', sepsis can be prevented by vaccinations against pneumococci and meningococci. Recently, the NIH NHLBI Panel redefined sepsis as 'severe endothelial dysfunction syndrome in response to intravascular and extravascular infections causing reversible or irreversible injury to the microcirculation responsible for multiple organ failure'. Microvascular endothelial injury underlies sepsis-associated hypotension, edema, disseminated intravascular coagulation, acute respiratory distress syndrome and acute kidney injury. Microbial genome products trigger 'genome wars' in sepsis that reprogram the human genome and culminate in a 'genomic storm' in blood and vascular cells. Sepsis can be averted experimentally by endothelial cytoprotection through targeting nuclear signaling that mediates inflammation and deranged metabolism. Endothelial 'rheostats' (e.g. inhibitors of NF-κB, A20 protein, CRADD/RAIDD protein and microRNAs) regulate endothelial signaling. Physiologic 'extinguishers' (e.g. suppressor of cytokine signaling 3) can be replenished through intracellular protein therapy. Lipid mediators (e.g. resolvin D1) hasten sepsis resolution. As sepsis cases rose from 387 330 in 1996 to 1.1 million in 2011, and are estimated to reach 2 million by 2020 in the US, mortality due to sepsis approaches that of heart attacks and exceeds deaths from stroke. More preventive vaccines and therapeutic measures are urgently needed.
Collapse
Affiliation(s)
- J Hawiger
- Immunotherapy Program at Vanderbilt University School of Medicine, Nashville, TN, USA
- Department of Medicine Division of Allergy Pulmonary and Critical Care Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - R A Veach
- Immunotherapy Program at Vanderbilt University School of Medicine, Nashville, TN, USA
- Department of Medicine Division of Allergy Pulmonary and Critical Care Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - J Zienkiewicz
- Immunotherapy Program at Vanderbilt University School of Medicine, Nashville, TN, USA
- Department of Medicine Division of Allergy Pulmonary and Critical Care Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| |
Collapse
|
348
|
Neves F, Abrantes J, Almeida T, de Matos AL, Costa PP, Esteves PJ. Genetic characterization of interleukins (IL-1α, IL-1β, IL-2, IL-4, IL-8, IL-10, IL-12A, IL-12B, IL-15 and IL-18) with relevant biological roles in lagomorphs. Innate Immun 2015; 21:787-801. [PMID: 26395994 PMCID: PMC4609935 DOI: 10.1177/1753425915606209] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 08/17/2015] [Indexed: 12/11/2022] Open
Abstract
ILs, as essential innate immune modulators, are involved in an array of biological processes. In the European rabbit (Oryctolagus cuniculus) IL-1α, IL-1β, IL-2, IL-4, IL-8, IL-10, IL-12A, IL-12B, IL-15 and IL-18 have been implicated in inflammatory processes and in the immune response against rabbit hemorrhagic disease virus and myxoma virus infections. In this study we characterized these ILs in six Lagomorpha species (European rabbit, pygmy rabbit, two cottontail rabbit species, European brown hare and American pika). Overall, these ILs are conserved between lagomorphs, including in their exon/intron structure. Most differences were observed between leporids and American pika. Indeed, when comparing both, some relevant differences were observed in American pika, such as the location of the stop codon in IL-1α and IL-2, the existence of a different transcript in IL8 and the number of cysteine residues in IL-1β. Changes at N-glycosylation motifs were also detected in IL-1, IL-10, IL-12B and IL-15. IL-1α is the protein that presents the highest evolutionary distances, which is in contrast to IL-12A where the distances between lagomorphs are the lowest. For all these ILs, sequences of human and European rabbit are more closely related than between human and mouse or European rabbit and mouse.
Collapse
Affiliation(s)
- Fabiana Neves
- CIBIO, InBIO-Research Network in Biodiversity and Evolutionary Biology, Universidade do Porto, Campus de Vairão, Vairão, Portugal UMIB/UP-Unidade Multidisciplinar de Investigação Biomédica/Universidade do Porto, Porto, Portugal
| | - Joana Abrantes
- CIBIO, InBIO-Research Network in Biodiversity and Evolutionary Biology, Universidade do Porto, Campus de Vairão, Vairão, Portugal
| | - Tereza Almeida
- CIBIO, InBIO-Research Network in Biodiversity and Evolutionary Biology, Universidade do Porto, Campus de Vairão, Vairão, Portugal
| | - Ana Lemos de Matos
- Department of Molecular Genetics and Microbiology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Paulo P Costa
- UMIB/UP-Unidade Multidisciplinar de Investigação Biomédica/Universidade do Porto, Porto, Portugal Departmento Genética, CSPGF, Instituto Nacional de Saúde Dr. Ricardo Jorge, Porto, Portugal
| | - Pedro J Esteves
- CIBIO, InBIO-Research Network in Biodiversity and Evolutionary Biology, Universidade do Porto, Campus de Vairão, Vairão, Portugal Departamento de Biologia, Faculdade de Ciências da Universidade do Porto, Porto, Portugal CITS-Centro de Investigação em Tecnologias de Saúde, CESPU, Gandra, Portugal
| |
Collapse
|
349
|
Iaconelli C, Lemetais G, Kechaou N, Chain F, Bermúdez-Humarán LG, Langella P, Gervais P, Beney L. Drying process strongly affects probiotics viability and functionalities. J Biotechnol 2015; 214:17-26. [PMID: 26325197 DOI: 10.1016/j.jbiotec.2015.08.022] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Revised: 06/22/2015] [Accepted: 08/25/2015] [Indexed: 11/19/2022]
Abstract
Probiotic formulations are widely used and are proposed to have a variety of beneficial effects, depending on the probiotic strains present in the product. The impact of drying processes on the viability of probiotics is well documented. However, the impact of these processes on probiotics functionality remains unclear. In this work, we investigated variations in seven different bacterial markers after various desiccation processes. Markers were composed of four different viability evaluation (combining two growth abilities and two cytometric measurements) and in three in vitro functionalities: stimulation of IL-10 and IL-12 production by PBMCs (immunomodulation) and bacterial adhesion to hexadecane. We measured the impact of three drying processes (air-drying, freeze-drying and spray-drying), without the use of protective agents, on three types of probiotic bacteria: Bifidobacterium bifidum, Lactobacillus plantarum and Lactobacillus zeae. Our results show that the bacteria respond differently to the three different drying processes, in terms of viability and functionality. Drying methods produce important variations in bacterial immunomodulation and hydrophobicity, which are correlated. We also show that adherence can be stimulated (air-drying) or inhibited (spray-drying) by drying processes. Results of a multivariate analysis show no direct correlation between bacterial survival and functionality, but do show a correlation between probiotic responses to desiccation-rewetting and the process used to dry the bacteria.
Collapse
Affiliation(s)
- Cyril Iaconelli
- UMR Procédés Alimentaires et Microbiologiques, Université de Bourgogne, AgroSup Dijon, 1 Esplanade Erasme, 21000 Dijon, France
| | - Guillaume Lemetais
- Merck Medication Familiale, 18C boulevard Winston Churchill, 21000 Dijon, France
| | - Noura Kechaou
- Institut MICALIS, UMR 1319, Domaine de Vilvert, 78352 Jouy en Josas, France
| | - Florian Chain
- Institut MICALIS, UMR 1319, Domaine de Vilvert, 78352 Jouy en Josas, France
| | | | - Philippe Langella
- Institut MICALIS, UMR 1319, Domaine de Vilvert, 78352 Jouy en Josas, France
| | - Patrick Gervais
- UMR Procédés Alimentaires et Microbiologiques, Université de Bourgogne, AgroSup Dijon, 1 Esplanade Erasme, 21000 Dijon, France
| | - Laurent Beney
- UMR Procédés Alimentaires et Microbiologiques, Université de Bourgogne, AgroSup Dijon, 1 Esplanade Erasme, 21000 Dijon, France.
| |
Collapse
|
350
|
Riese P, Trittel S, Schulze K, Guzmán CA. Rodents as pre-clinical models for predicting vaccine performance in humans. Expert Rev Vaccines 2015. [DOI: 10.1586/14760584.2015.1074043] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
| | - Stephanie Trittel
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - Kai Schulze
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - Carlos A Guzmán
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| |
Collapse
|