1
|
Becker S, Allen J, Morison ZL, Saeid S, Adderley A, Koskelainen A, Vinberg F. Healing of ischemic injury in the retina. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.11.04.621932. [PMID: 39574566 PMCID: PMC11580909 DOI: 10.1101/2024.11.04.621932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/05/2025]
Abstract
Neuro- and retinal degenerative diseases, including Alzheimer's, age-related macular degeneration, stroke, and central retinal artery occlusion, rob millions of their independence. Studying these diseases in human retinas has been hindered by the immediate loss of neuronal activity postmortem. While recent studies restored limited activity in postmortem CNS tissues, synchronized neuronal transmission >30 minutes postmortem remained elusive. Our study overcomes this barrier by reviving and sustaining light signal transmission in human retinas recovered up to four hours and stored 48 hours postmortem. We also establish infrared-based ex vivo imaging for precise sampling, a closed perfusion system for drug testing, and an ex vivo ischemia-reperfusion model in mouse and human retina. This platform enables testing of neuroprotective and neurotoxic effects of drugs targeting oxidative stress and glutamate excitotoxicity. Our advances question the irreversibility of ischemic injury, support preclinical vision restoration studies, offer new insights into treating ischemic CNS injuries, and pave the way for transplantation of human donor eyes. Teaser Reviving light signaling in postmortem human retinas challenges the irreversibility of ischemic injury and advances research to restore vision.
Collapse
|
2
|
Kastner AL, Marx AF, Dimitrova M, Abreu-Mota T, Ertuna YI, Bonilla WV, Stauffer K, Künzli M, Wagner I, Kreutzfeldt M, Merkler D, Pinschewer DD. Durable lymphocyte subset elimination upon a single dose of AAV-delivered depletion antibody dissects immune control of chronic viral infection. Immunity 2025; 58:481-498.e10. [PMID: 39719711 DOI: 10.1016/j.immuni.2024.11.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 10/02/2024] [Accepted: 11/26/2024] [Indexed: 12/26/2024]
Abstract
To interrogate the role of specific immune cells in infection, cancer, and autoimmunity, immunologists commonly use monoclonal depletion antibodies (depletion-mAbs) or genetically engineered mouse models (GEMMs). To generate a tool that combines specific advantages and avoids select drawbacks of the two methods, we engineered adeno-associated viral vectors expressing depletion mAbs (depletion-AAVs). Single-dose depletion-AAV administration durably eliminated lymphocyte subsets in mice and avoided accessory deficiencies of GEMMs, such as marginal zone defects in B cell-deficient animals. Depletion-AAVs can be used in animals of different genetic backgrounds, and multiple depletion-AAVs can readily be combined. Exploiting depletion-AAV technology, we showed that B cells were required for unimpaired CD4+ and CD8+ T cell responses to chronic lymphocytic choriomeningitis virus (LCMV) infection. Upon B cell depletion, CD8+ T cells failed to suppress viremia, and they only helped resolve chronic infection when antibodies dampened viral loads. Our study positions depletion-AAVs as a versatile tool for immunological research.
Collapse
Affiliation(s)
- Anna Lena Kastner
- Department of Biomedicine, University of Basel, 4009 Basel, Switzerland
| | | | - Mirela Dimitrova
- Department of Biomedicine, University of Basel, 4009 Basel, Switzerland
| | - Tiago Abreu-Mota
- Department of Biomedicine, University of Basel, 4009 Basel, Switzerland
| | - Yusuf I Ertuna
- Department of Biomedicine, University of Basel, 4009 Basel, Switzerland
| | - Weldy V Bonilla
- Department of Biomedicine, University of Basel, 4009 Basel, Switzerland
| | - Karsten Stauffer
- Department of Biomedicine, University of Basel, 4009 Basel, Switzerland
| | - Marco Künzli
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., 4070 Basel, Switzerland
| | - Ingrid Wagner
- Department of Pathology and Immunology, University of Geneva, 1211 Geneva, Switzerland
| | - Mario Kreutzfeldt
- Department of Pathology and Immunology, University of Geneva, 1211 Geneva, Switzerland; Division of Clinical Pathology, Geneva University Hospital, 1206 Geneva, Switzerland
| | - Doron Merkler
- Department of Pathology and Immunology, University of Geneva, 1211 Geneva, Switzerland; Division of Clinical Pathology, Geneva University Hospital, 1206 Geneva, Switzerland
| | | |
Collapse
|
3
|
Vinogradova TM, Lakatta EG. Ca 2+/Calmodulin-Dependent Protein Kinase II (CaMKII) Regulates Basal Cardiac Pacemaker Function: Pros and Cons. Cells 2024; 14:3. [PMID: 39791704 PMCID: PMC11719954 DOI: 10.3390/cells14010003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Revised: 12/14/2024] [Accepted: 12/21/2024] [Indexed: 01/12/2025] Open
Abstract
The spontaneous firing of the sinoatrial (SA) node, the physiological pacemaker of the heart, is generated within sinoatrial nodal cells (SANCs) and is regulated by a "coupled-clock" pacemaker system, which integrates a "membrane clock", the ensemble of ion channel currents, and an intracellular "Ca2+ clock", sarcoplasmic reticulum-generated local submembrane Ca2+ releases via ryanodine receptors. The interactions within a "coupled-clock" system are modulated by phosphorylation of surface membrane and sarcoplasmic reticulum proteins. Though the essential role of a high basal cAMP level and PKA-dependent phosphorylation for basal spontaneous SANC firing is well recognized, the role of basal CaMKII-dependent phosphorylation remains uncertain. This is a critical issue with respect to how cardiac pacemaker cells fire spontaneous action potentials. This review aspires to explain and unite apparently contradictory results of pharmacological studies in the literature that have demonstrated a fundamental role of basal CaMKII activation for basal cardiac pacemaker function, as well as studies in mice with genetic CaMKII inhibition which have been interpreted to indicate that basal spontaneous SANC firing is independent of CaMKII activation. The assessment of supporting and opposing data regarding CaMKII effects on phosphorylation of Ca2+-cycling proteins and spontaneous firing of SANC in the basal state leads to the necessary conclusion that CaMKII activity and CaMKII-dependent phosphorylation do regulate basal cardiac pacemaker function.
Collapse
Affiliation(s)
- Tatiana M. Vinogradova
- Laboratory of Cardiovascular Science, Intramural Research Program, National Institute on Aging, National Institute of Health, Baltimore, MD 21224, USA;
| | | |
Collapse
|
4
|
Matye D, Leak J, Woolbright BL, Taylor JA. Preclinical models of bladder cancer: BBN and beyond. Nat Rev Urol 2024; 21:723-734. [PMID: 38769130 DOI: 10.1038/s41585-024-00885-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/10/2024] [Indexed: 05/22/2024]
Abstract
Preclinical modelling is a crucial component of advancing the understanding of cancer biology and therapeutic development. Several models exist for understanding the pathobiology of bladder cancer and evaluating therapeutics. N-butyl-N-(4-hydroxybutyl)-nitrosamine (BBN)-induced bladder cancer is a commonly used model that recapitulates many of the features of human disease. Particularly in mice, BBN is a preferred laboratory model owing to a high level of reproducibility, high genetic fidelity to the human condition, and its relative ease of use. However, important aspects of the model are often overlooked in laboratory studies. Moreover, the advent of new models has yielded a variety of methodologies that complement the use of BBN. Toxicokinetics, histopathology, molecular genetics and sex can differ between available models and are important factors to consider in bladder cancer modelling.
Collapse
Affiliation(s)
- David Matye
- School of Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Juliann Leak
- School of Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Benjamin L Woolbright
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS, USA
| | - John A Taylor
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS, USA.
- Department of Urology, University of Kansas Medical Center, Kansas City, KS, USA.
| |
Collapse
|
5
|
Paz HA, Buddha L, Zhong Y, Sikes JD, Wankhade UD. Impact of maternal high-fat diet on offspring gut microbiota during short-term high-fat diet exposure in mice. Physiol Rep 2024; 12:e70111. [PMID: 39489538 PMCID: PMC11531878 DOI: 10.14814/phy2.70111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 10/16/2024] [Accepted: 10/16/2024] [Indexed: 11/05/2024] Open
Abstract
Alterations in the gut microbiome have been linked to obesity, with maternal high-fat diet (HF) playing a role in shaping offspring microbiome composition. However, the sex-specific responses to maternal HF diet and the impact of subsequent dietary challenges remain unclear. This study investigated the effects of maternal HF diet on offspring gut microbiota structure and predicted functional profile in response to short-term postnatal HF diet exposure with a focus on sex-specific responses. Female and male offspring of maternal control (C) diet or maternal HF diet were weaned onto C diet or HF diet. Offspring were euthanized at 13 weeks of age and cecal contents were collected for bacterial taxonomic profiling. Maternal HF diet reduced α-diversity, notably in male offspring weaned onto HF diet. Sex-specific differences were observed in the gut microbial composition and predicted functional potential. Furthermore, the influence of maternal diet on bacterial community structure and functional potential varied depending on postnatal diet. Maternal HF diet led to increased relative abundance of Corynebacterium in female offspring and decreased abundance of Akkermansia and Roseburia in male offspring. These findings underscore the sexually dimorphic nature of maternal HF diet effects on gut microbiota composition and function, with implications for developmental programming and metabolic health.
Collapse
Affiliation(s)
- Henry A. Paz
- Department of Pediatrics, College of MedicineUniversity of Arkansas for Medical SciencesLittle RockArkansasUSA
- Arkansas Children's Nutrition CenterLittle RockArkansasUSA
| | - Lasya Buddha
- Department of Pediatrics, College of MedicineUniversity of Arkansas for Medical SciencesLittle RockArkansasUSA
| | - Ying Zhong
- Department of Pediatrics, College of MedicineUniversity of Arkansas for Medical SciencesLittle RockArkansasUSA
- Arkansas Children's Nutrition CenterLittle RockArkansasUSA
| | - James D. Sikes
- Arkansas Children's Nutrition CenterLittle RockArkansasUSA
| | - Umesh D. Wankhade
- Department of Pediatrics, College of MedicineUniversity of Arkansas for Medical SciencesLittle RockArkansasUSA
- Arkansas Children's Nutrition CenterLittle RockArkansasUSA
| |
Collapse
|
6
|
Andrade F, Howell L, Percival CJ, Richtsmeier JT, Marcucio RS, Hallgrímsson B, Cheverud JM. Genetic architecture of trait variance in craniofacial morphology. Genetics 2024; 226:iyae028. [PMID: 38386896 PMCID: PMC11090463 DOI: 10.1093/genetics/iyae028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 12/19/2023] [Accepted: 01/26/2024] [Indexed: 02/24/2024] Open
Abstract
The genetic architecture of trait variance has long been of interest in genetics and evolution. One of the earliest attempts to understand this architecture was presented in Lerner's Genetic Homeostasis (1954). Lerner proposed that heterozygotes should be better able to tolerate environmental perturbations because of functional differences between the alleles at a given locus, with each allele optimal for slightly different environments. This greater robustness to environmental variance, he argued, would result in smaller trait variance for heterozygotes. The evidence for Lerner's hypothesis has been inconclusive. To address this question using modern genomic methods, we mapped loci associated with differences in trait variance (vQTL) on 1,101 individuals from the F34 of an advanced intercross between LG/J and SM/J mice. We also mapped epistatic interactions for these vQTL in order to understand the influence of epistasis for the architecture of trait variance. We did not find evidence supporting Lerner's hypothesis, that heterozygotes tend to have smaller trait variances than homozygotes. We further show that the effects of most mapped loci on trait variance are produced by epistasis affecting trait means and that those epistatic effects account for about a half of the differences in genotypic-specific trait variances. Finally, we propose a model where the different interactions between the additive and dominance effects of the vQTL and their epistatic partners can explain Lerner's original observations but can also be extended to include other conditions where heterozygotes are not the least variable genotype.
Collapse
Affiliation(s)
- Fernando Andrade
- Department of Biology, Loyola University Chicago, Chicago, IL 60660, USA
| | - Lisa Howell
- Department of Anthropology, Penn State University, University Park, PA 16802, USA
| | | | - Joan T Richtsmeier
- Department of Anthropology, Penn State University, University Park, PA 16802, USA
| | - Ralph S Marcucio
- Department of Orthopedic Surgery, School of Medicine, University of California San Francisco, San Francisco, CA 94110, USA
| | - Benedikt Hallgrímsson
- Department of Cell Biology and Anatomy, Cumming School of Medicine, Alberta Children's Hospital Research Institute and McCaig Bone and Joint Institute, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - James M Cheverud
- Department of Biology, Loyola University Chicago, Chicago, IL 60660, USA
| |
Collapse
|
7
|
Chu JTS, Lamers MM. Organoids in virology. NPJ VIRUSES 2024; 2:5. [PMID: 40295690 PMCID: PMC11721363 DOI: 10.1038/s44298-024-00017-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 01/04/2024] [Indexed: 04/30/2025]
Abstract
To adequately prepare against imminent disease outbreaks from diverse and ever-changing viral pathogens, improved experimental models that can accurately recapitulate host-virus responses and disease pathogenesis in human are essential. Organoid platforms have emerged in recent years as amenable in vitro tools that can bridge the limitations of traditional 2D cell lines and animal models for viral disease research. We highlight in this review the key insights that have contributed by organoid models to virus research, the limitations that exist in current platforms, and outline novel approaches that are being applied to address these shortcomings.
Collapse
Affiliation(s)
- Julie T S Chu
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
| | - Mart M Lamers
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore.
| |
Collapse
|
8
|
Chun T, Pattem J, Gillis RB, Dinu VT, Yakubov GE, Corfield AP, Harding SE. Comparative hydrodynamic and nanoscale imaging study on the interactions of teicoplanin-A2 and bovine submaxillary mucin as a model ocular mucin. Sci Rep 2023; 13:11367. [PMID: 37443326 PMCID: PMC10344913 DOI: 10.1038/s41598-023-38036-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 07/01/2023] [Indexed: 07/15/2023] Open
Abstract
Glycopeptide antibiotics are regularly used in ophthalmology to treat infections of Gram-positive bacteria. Aggregative interactions of antibiotics with mucins however can lead to long exposure and increases the risk of resistant species. This study focuses on the evaluation of potential interactions of the last line of defence glycopeptide antibiotic teicoplanin with an ocular mucin model using precision matrix free hydrodynamic and microscopic techniques: sedimentation velocity in the analytical ultracentrifuge (SV-AUC), dynamic light scattering (DLS) and atomic force microscopy (AFM). For the mixtures of teicoplanin at higher doses (1.25 mg/mL and 12.5 mg/mL), it was shown to interact and aggregate with bovine submaxillary mucin (BSM) in the distributions of both sedimentation coefficients by SV-AUC and hydrodynamic radii by DLS. The presence of aggregates was confirmed by AFM for higher concentrations. We suggest that teicoplanin eye drop formulations should be delivered at concentrations of < 1.25 mg/mL to avoid potentially harmful aggregations.
Collapse
Affiliation(s)
- Taewoo Chun
- National Centre for Macromolecular Hydrodynamics, University of Nottingham, Sutton Bonington, LE12 5RD, UK
| | - Jacob Pattem
- National Centre for Macromolecular Hydrodynamics, University of Nottingham, Sutton Bonington, LE12 5RD, UK
- Soft Matter Biomaterials and Biointerfaces, School of Biosciences, University of Nottingham, Sutton Bonington, LE12 5RD, UK
| | - Richard B Gillis
- College of Business, Technology and Engineering, Food and Nutrition Group, Sheffield Hallam University, Arundel Gate, Sheffield, S1 1WB, UK
| | - Vlad T Dinu
- National Centre for Macromolecular Hydrodynamics, University of Nottingham, Sutton Bonington, LE12 5RD, UK
| | - Gleb E Yakubov
- Soft Matter Biomaterials and Biointerfaces, School of Biosciences, University of Nottingham, Sutton Bonington, LE12 5RD, UK
| | - Anthony P Corfield
- National Centre for Macromolecular Hydrodynamics, University of Nottingham, Sutton Bonington, LE12 5RD, UK
| | - Stephen E Harding
- National Centre for Macromolecular Hydrodynamics, University of Nottingham, Sutton Bonington, LE12 5RD, UK.
| |
Collapse
|
9
|
Patel P, Nandi A, Verma SK, Kaushik N, Suar M, Choi EH, Kaushik NK. Zebrafish-based platform for emerging bio-contaminants and virus inactivation research. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 872:162197. [PMID: 36781138 PMCID: PMC9922160 DOI: 10.1016/j.scitotenv.2023.162197] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/23/2023] [Accepted: 02/08/2023] [Indexed: 05/27/2023]
Abstract
Emerging bio-contaminants such as viruses have affected health and environment settings of every country. Viruses are the minuscule entities resulting in severe contagious diseases like SARS, MERS, Ebola, and avian influenza. Recent epidemic like the SARS-CoV-2, the virus has undergone mutations strengthen them and allowing to escape from the remedies. Comprehensive knowledge of viruses is essential for the development of targeted therapeutic and vaccination treatments. Animal models mimicking human biology like non-human primates, rats, mice, and rabbits offer competitive advantage to assess risk of viral infections, chemical toxins, nanoparticles, and microbes. However, their economic maintenance has always been an issue. Furthermore, the redundancy of experimental results due to aforementioned aspects is also in examine. Hence, exploration for the alternative animal models is crucial for risk assessments. The current review examines zebrafish traits and explores the possibilities to monitor emerging bio-contaminants. Additionally, a comprehensive picture of the bio contaminant and virus particle invasion and abatement mechanisms in zebrafish and human cells is presented. Moreover, a zebrafish model to investigate the emerging viruses such as coronaviridae and poxviridae has been suggested.
Collapse
Affiliation(s)
- Paritosh Patel
- Plasma Bioscience Research Center, Department of Electrical and Biological Physics, Kwangwoon University, 01897 Seoul, South Korea
| | - Aditya Nandi
- School of Biotechnology, KIIT University, Bhubaneswar 751024, Odisha, India
| | - Suresh K Verma
- School of Biotechnology, KIIT University, Bhubaneswar 751024, Odisha, India; Condensed Matter Theory Group, Materials Theory Division, Department of Physics and Astronomy, Uppsala University, Box 516, SE-751 20 Uppsala, Sweden
| | - Neha Kaushik
- Department of Biotechnology, College of Engineering, The University of Suwon, 18323 Hwaseong, Republic of Korea
| | - Mrutyunjay Suar
- School of Biotechnology, KIIT University, Bhubaneswar 751024, Odisha, India
| | - Eun Ha Choi
- Plasma Bioscience Research Center, Department of Electrical and Biological Physics, Kwangwoon University, 01897 Seoul, South Korea.
| | - Nagendra Kumar Kaushik
- Plasma Bioscience Research Center, Department of Electrical and Biological Physics, Kwangwoon University, 01897 Seoul, South Korea.
| |
Collapse
|
10
|
Terrones M, de Beeck KO, Van Camp G, Vandeweyer G. Pre-clinical modelling of ROS1+ non-small cell lung cancer. Lung Cancer 2023; 180:107192. [PMID: 37068393 DOI: 10.1016/j.lungcan.2023.107192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 04/04/2023] [Accepted: 04/08/2023] [Indexed: 04/19/2023]
Abstract
Non-small cell lung cancer (NSCLC) is a heterogeneous group of diseases which accounts for 80% of newly diagnosed lung cancers. In the previous decade, a new molecular subset of NSCLC patients (around 2%) harboring rearrangements of the c-ros oncogene 1 was defined. ROS1+ NSCLC is typically diagnosed in young, nonsmoker individuals presenting an adenocarcinoma histology. Patients can benefit from tyrosine kinase inhibitors (TKIs) such as crizotinib and entrectinib, compounds initially approved to treat ALK-, MET- or NTRK- rearranged malignancies respectively. Given the low prevalence of ROS1-rearranged tumors, the use of TKIs was authorized based on pre-clinical evidence using limited experimental models, followed by basket clinical trials. After initiating targeted therapy, disease relapse is reported in approximately 50% of cases as a result of the appearance of resistance mechanisms. The restricted availability of TKIs active against resistance events critically reduces the overall survival. In this review we discuss the pre-clinical ROS1+ NSCLC models developed up to date, highlighting their strengths and limitations with respect to the unmet clinical needs. By combining gene-editing tools and novel cell culture approaches, newly developed pre-clinical models will enhance the development of next-generation tyrosine kinase inhibitors that overcome resistant tumor cell subpopulations.
Collapse
Affiliation(s)
- Marc Terrones
- Center of Medical Genetics, University of Antwerp and Antwerp University Hospital, Prins Boudewijnlaan 43/6, 2650 Edegem, Belgium; Center for Oncological Research, University of Antwerp and Antwerp University Hospital, Universiteitsplein 1, 2610 Wilrijk, Belgium.
| | - Ken Op de Beeck
- Center of Medical Genetics, University of Antwerp and Antwerp University Hospital, Prins Boudewijnlaan 43/6, 2650 Edegem, Belgium; Center for Oncological Research, University of Antwerp and Antwerp University Hospital, Universiteitsplein 1, 2610 Wilrijk, Belgium
| | - Guy Van Camp
- Center of Medical Genetics, University of Antwerp and Antwerp University Hospital, Prins Boudewijnlaan 43/6, 2650 Edegem, Belgium; Center for Oncological Research, University of Antwerp and Antwerp University Hospital, Universiteitsplein 1, 2610 Wilrijk, Belgium
| | - Geert Vandeweyer
- Center of Medical Genetics, University of Antwerp and Antwerp University Hospital, Prins Boudewijnlaan 43/6, 2650 Edegem, Belgium
| |
Collapse
|
11
|
Hwang S, Ha Y, Koo G, Noh H, Lee A, Kim B, Hong SM, Morgan MJ, Eyun S, Lee D, Roe J, Lee Y, Kim Y. LCK-Mediated RIPK3 Activation Controls Double-Positive Thymocyte Proliferation and Restrains Thymic Lymphoma by Regulating the PP2A-ERK Axis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2204522. [PMID: 36161785 PMCID: PMC9661840 DOI: 10.1002/advs.202204522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 09/07/2022] [Indexed: 06/16/2023]
Abstract
Receptor-interacting protein kinase 3 (RIPK3) is the primary regulator of necroptotic cell death. RIPK3 expression is often silenced in various cancer cells, which suggests that it may have tumor suppressor properties. However, the exact mechanism by which RIPK3 negatively regulates cancer development and progression remains unclear. This report indicates that RIPK3 acts as a potent regulator of the homeostatic proliferation of CD4+ CD8+ double-positive (DP) thymocytes. Abnormal proliferation of RIPK3-deficient DP thymocytes occurs independently of the well-known role for RIPK3 in necroptosis (upstream of MLKL activation), and is associated with an incidental thymic mass, likely thymic hyperplasia. In addition, Ripk3-null mice develop increased thymic tumor formation accompanied by reduced host survival in the context of an N-ethyl-N-nitrosourea (ENU)-induced tumor model. Moreover, RIPK3 deficiency in p53-null mice promotes thymic lymphoma development via upregulated extracellular signal-regulated kinase (ERK) signaling, which correlates with markedly reduced survival rates. Mechanistically, lymphocyte-specific protein tyrosine kinase (LCK) activates RIPK3, which in turn leads to increases in the phosphatase activity of protein phosphatase 2 (PP2A), thereby suppressing hyper-activation of ERK in DP thymocytes. Overall, these findings suggest that a RIPK3-PP2A-ERK signaling axis regulates DP thymocyte homeostasis and may provide a potential therapeutic target to improve thymic lymphoma therapies.
Collapse
Affiliation(s)
- Sung‐Min Hwang
- Department of Biochemistry and Molecular BiologyAjou University School of Medicine164 Worldcup‐ro, Yeongtong‐gu, SuwonGyeonggi‐do16499Republic of Korea
- Sandra and Edward Meyer Cancer Center and Department of Obstetrics and GynecologyWeill Cornell MedicineNew YorkNY10065USA
| | - Yu‐Jin Ha
- Department of Biochemistry and Molecular BiologyAjou University School of Medicine164 Worldcup‐ro, Yeongtong‐gu, SuwonGyeonggi‐do16499Republic of Korea
- Department of Biomedical SciencesGraduate School of Ajou University164 Worldcup‐ro, Yeongtong‐gu, SuwonGyeonggi‐do16499Republic of Korea
| | - Gi‐Bang Koo
- Department of Biochemistry and Molecular BiologyAjou University School of Medicine164 Worldcup‐ro, Yeongtong‐gu, SuwonGyeonggi‐do16499Republic of Korea
- Department of Biomedical SciencesGraduate School of Ajou University164 Worldcup‐ro, Yeongtong‐gu, SuwonGyeonggi‐do16499Republic of Korea
| | - Hyun‐Jin Noh
- Department of Biochemistry and Molecular BiologyAjou University School of Medicine164 Worldcup‐ro, Yeongtong‐gu, SuwonGyeonggi‐do16499Republic of Korea
- Department of Biomedical SciencesGraduate School of Ajou University164 Worldcup‐ro, Yeongtong‐gu, SuwonGyeonggi‐do16499Republic of Korea
| | - A‐Yeon Lee
- Department of Biochemistry and Molecular BiologyAjou University School of Medicine164 Worldcup‐ro, Yeongtong‐gu, SuwonGyeonggi‐do16499Republic of Korea
- Department of Biomedical SciencesGraduate School of Ajou University164 Worldcup‐ro, Yeongtong‐gu, SuwonGyeonggi‐do16499Republic of Korea
| | - Byeong‐Ju Kim
- Department of Biochemistry and Molecular BiologyAjou University School of Medicine164 Worldcup‐ro, Yeongtong‐gu, SuwonGyeonggi‐do16499Republic of Korea
- Department of Biomedical SciencesGraduate School of Ajou University164 Worldcup‐ro, Yeongtong‐gu, SuwonGyeonggi‐do16499Republic of Korea
| | - Sun Mi Hong
- Department of Biochemistry and Molecular BiologyAjou University School of Medicine164 Worldcup‐ro, Yeongtong‐gu, SuwonGyeonggi‐do16499Republic of Korea
| | - Michael J. Morgan
- Department of Natural SciencesNortheastern State UniversityTahlequahOK74464USA
| | - Seong‐il Eyun
- Department of Life ScienceChung‐Ang UniversitySeoul06973Republic of Korea
| | - Dakeun Lee
- Department of PathologyAjou University School of Medicine164 Worldcup‐ro, Yeongtong‐gu, SuwonGyeonggi‐do16499Republic of Korea
| | - Jae‐Seok Roe
- Department of BiochemistryCollege of Life Science and BiotechnologyYonsei UniversitySeoul03722Republic of Korea
| | - Youngsoo Lee
- Institute of Medical ScienceAjou University School of Medicine164 Worldcup‐ro, Yeongtong‐gu, SuwonGyeonggi‐do16499Republic of Korea
| | - You‐Sun Kim
- Department of Biochemistry and Molecular BiologyAjou University School of Medicine164 Worldcup‐ro, Yeongtong‐gu, SuwonGyeonggi‐do16499Republic of Korea
- Department of Biomedical SciencesGraduate School of Ajou University164 Worldcup‐ro, Yeongtong‐gu, SuwonGyeonggi‐do16499Republic of Korea
| |
Collapse
|
12
|
Dolan B, Ermund A, Martinez-Abad B, Johansson ME, Hansson GC. Clearance of small intestinal crypts involves goblet cell mucus secretion by intracellular granule rupture and enterocyte ion transport. Sci Signal 2022; 15:eabl5848. [PMID: 36126118 PMCID: PMC9749883 DOI: 10.1126/scisignal.abl5848] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Goblet cells in the small intestinal crypts contain large numbers of mucin granules that are rapidly discharged to clean bacteria from the crypt. Because acetylcholine released by neuronal and nonneuronal cells controls many aspects of intestinal epithelial function, we used tissue explants and organoids to investigate the response of the small intestinal crypt to cholinergic stimulation. The activation of muscarinic acetylcholine receptors initiated a coordinated and rapid emptying of crypt goblet cells that flushed the crypt contents into the intestinal lumen. Cholinergic stimulation induced an expansion of the granule contents followed by intracellular rupture of the mucin granules. The mucus expanded intracellularly before the rupture of the goblet cell apical membrane and continued to expand after its release into the crypt lumen. The goblet cells recovered from membrane rupture and replenished their stores of mucin granules. Mucus secretion from the goblet cells depended on Ca2+ signaling and the expansion of the mucus in the crypt depended on gap junctions and on ion and water transport by enterocytes adjacent to the goblet cells. This distinctive mode of mucus secretion, which we refer to as "expanding secretion," efficiently cleans the small intestine crypt through coordinated mucus, ion, and fluid secretion by goblet cells and enterocytes.
Collapse
Affiliation(s)
- Brendan Dolan
- Department of Medical Biochemistry and Cell Biology, University of
Gothenburg, 405 30 Gothenburg, Sweden
| | - Anna Ermund
- Department of Medical Biochemistry and Cell Biology, University of
Gothenburg, 405 30 Gothenburg, Sweden
| | - Beatriz Martinez-Abad
- Department of Medical Biochemistry and Cell Biology, University of
Gothenburg, 405 30 Gothenburg, Sweden
| | - Malin E.V. Johansson
- Department of Medical Biochemistry and Cell Biology, University of
Gothenburg, 405 30 Gothenburg, Sweden
| | - Gunnar C. Hansson
- Department of Medical Biochemistry and Cell Biology, University of
Gothenburg, 405 30 Gothenburg, Sweden
| |
Collapse
|
13
|
Hosur V, Low BE, Wiles MV. Programmable RNA-Guided Large DNA Transgenesis by CRISPR/Cas9 and Site-Specific Integrase Bxb1. Front Bioeng Biotechnol 2022; 10:910151. [PMID: 35866031 PMCID: PMC9294445 DOI: 10.3389/fbioe.2022.910151] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 06/01/2022] [Indexed: 11/17/2022] Open
Abstract
The inability to insert large DNA constructs into the genome efficiently and precisely is a key challenge in genomic engineering. Random transgenesis, which is widely used, lacks precision, and comes with a slew of drawbacks. Lentiviral and adeno-associated viral methods are plagued by, respectively, DNA toxicity and a payload capacity of less than 5 kb. Homology-directed repair (HDR) techniques based on CRISPR-Cas9 can be effective, but only in the 1-5 kb range. In addition, long homology arms-DNA sequences that permit construct insertion-of lengths ranging from 0.5 to 5 kb are required by currently known HDR-based techniques. A potential new method that uses Cas9-guided transposases to insert DNA structures up to 10 kb in length works well in bacteria, but only in bacteria. Surmounting these roadblocks, a new toolkit has recently been developed that combines RNA-guided Cas9 and the site-specific integrase Bxb1 to integrate DNA constructs ranging in length from 5 to 43 kb into mouse zygotes with germline transmission and into human cells. This ground-breaking toolkit will give researchers a valuable resource for developing novel, urgently needed mouse and human induced pluripotent stem cell (hiPSC) models of cancer and other genetic diseases, as well as therapeutic gene integration and biopharmaceutical applications, such as the development of stable cell lines to produce therapeutic protein products.
Collapse
Affiliation(s)
- Vishnu Hosur
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME, United States
| | | | | |
Collapse
|
14
|
Iglesias-Carres L, Neilson AP. Utilizing preclinical models of genetic diversity to improve translation of phytochemical activities from rodents to humans and inform personalized nutrition. Food Funct 2021; 12:11077-11105. [PMID: 34672309 DOI: 10.1039/d1fo02782d] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Mouse models are an essential tool in different areas of research, including nutrition and phytochemical research. Traditional inbred mouse models have allowed the discovery of therapeutical targets and mechanisms of action and expanded our knowledge of health and disease. However, these models lack the genetic variability typically found in human populations, which hinders the translatability of the results found in mice to humans. The development of genetically diverse mouse models, such as the collaborative cross (CC) or the diversity outbred (DO) models, has been a useful tool to overcome this obstacle in many fields, such as cancer, immunology and toxicology. However, these tools have not yet been widely adopted in the field of phytochemical research. As demonstrated in other disciplines, use of CC and DO models has the potential to provide invaluable insights for translation of phytochemicals from rodents to humans, which are desperately needed given the challenges and numerous failed clinical trials in this field. These models may prove informative for personalized use of phytochemicals in humans, including: predicting interindividual variability in phytochemical bioavailability and efficacy, identifying genetic loci or genes governing response to phytochemicals, identifying phytochemical mechanisms of action and therapeutic targets, and understanding the impact of genetic variability on individual response to phytochemicals. Such insights would prove invaluable for personalized implementation of phytochemicals in humans. This review will focus on the current work performed with genetically diverse mouse populations, and the research opportunities and advantages that these models can offer to phytochemical research.
Collapse
Affiliation(s)
- Lisard Iglesias-Carres
- Plants for Human Health Institute, Department of Food, Bioprocessing and Nutrition Sciences, North Carolina State University, Kannapolis, NC, USA.
| | - Andrew P Neilson
- Plants for Human Health Institute, Department of Food, Bioprocessing and Nutrition Sciences, North Carolina State University, Kannapolis, NC, USA.
| |
Collapse
|
15
|
Fiege JK, Block KE, Pierson MJ, Nanda H, Shepherd FK, Mickelson CK, Stolley JM, Matchett WE, Wijeyesinghe S, Meyerholz DK, Vezys V, Shen SS, Hamilton SE, Masopust D, Langlois RA. Mice with diverse microbial exposure histories as a model for preclinical vaccine testing. Cell Host Microbe 2021; 29:1815-1827.e6. [PMID: 34731647 DOI: 10.1016/j.chom.2021.10.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 08/30/2021] [Accepted: 10/08/2021] [Indexed: 12/13/2022]
Abstract
Laboratory mice comprise an expeditious model for preclinical vaccine testing; however, vaccine immunogenicity in these models often inadequately translates to humans. Reconstituting physiologic microbial experience to specific pathogen-free (SPF) mice induces durable immunological changes that better recapitulate human immunity. We examined whether mice with diverse microbial experience better model human responses post vaccination. We co-housed laboratory mice with pet-store mice, which have varied microbial exposures, and then assessed immune responses to influenza vaccines. Human transcriptional responses to influenza vaccination are better recapitulated in co-housed mice. Although SPF and co-housed mice were comparably susceptible to acute influenza infection, vaccine-induced humoral responses were dampened in co-housed mice, resulting in poor control upon challenge. Additionally, protective heterosubtypic T cell immunity was compromised in co-housed mice. Because SPF mice exaggerated humoral and T cell protection upon influenza vaccination, reconstituting microbial experience in laboratory mice through co-housing may better inform preclinical vaccine testing.
Collapse
Affiliation(s)
- Jessica K Fiege
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN 55455, USA; Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Katharine E Block
- Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA; Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Mark J Pierson
- Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA; Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Hezkiel Nanda
- Institute for Health Informatics, University of Minnesota, Minneapolis, MN 55455, USA
| | - Frances K Shepherd
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN 55455, USA; Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Clayton K Mickelson
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN 55455, USA; Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA
| | - J Michael Stolley
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN 55455, USA; Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA
| | - William E Matchett
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN 55455, USA; Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Sathi Wijeyesinghe
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN 55455, USA; Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA
| | - David K Meyerholz
- Department of Pathology, University of Iowa, Iowa City, IA 52242, USA
| | - Vaiva Vezys
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN 55455, USA; Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Steven S Shen
- Institute for Health Informatics, University of Minnesota, Minneapolis, MN 55455, USA
| | - Sara E Hamilton
- Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA; Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA.
| | - David Masopust
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN 55455, USA; Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA.
| | - Ryan A Langlois
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN 55455, USA; Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
16
|
van Daal MT, Folkerts G, Garssen J, Braber S. Pharmacological Modulation of Immune Responses by Nutritional Components. Pharmacol Rev 2021; 73:198-232. [PMID: 34663688 DOI: 10.1124/pharmrev.120.000063] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The incidence of noncommunicable diseases (NCDs) has increased over the last few decades, and one of the major contributors to this is lifestyle, especially diet. High intake of saturated fatty acids and low intake of dietary fiber is linked to an increase in NCDs. Conversely, a low intake of saturated fatty acids and a high intake of dietary fiber seem to have a protective effect on general health. Several mechanisms have been identified that underlie this phenomenon. In this review, we focus on pharmacological receptors, including the aryl hydrocarbon receptor, binding partners of the retinoid X receptor, G-coupled protein receptors, and toll-like receptors, which can be activated by nutritional components and their metabolites. Depending on the nutritional component and the receptors involved, both proinflammatory and anti-inflammatory effects occur, leading to an altered immune response. These insights may provide opportunities for the prevention and treatment of NCDs and their inherent (sub)chronic inflammation. SIGNIFICANCE STATEMENT: This review summarizes the reported effects of nutritional components and their metabolites on the immune system through manipulation of specific (pharmacological) receptors, including the aryl hydrocarbon receptor, binding partners of the retinoid X receptor, G-coupled protein receptors, and toll-like receptors. Nutritional components, such as vitamins, fibers, and unsaturated fatty acids are able to resolve inflammation, whereas saturated fatty acids tend to exhibit proinflammatory effects. This may aid decision makers and scientists in developing strategies to decrease the incidence of noncommunicable diseases.
Collapse
Affiliation(s)
- Marthe T van Daal
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584 CG, Utrecht, The Netherlands (M.T.v.D., G.F., J.G., S.B.); and Danone Nutricia Research, 3584 CT, Utrecht, The Netherlands (J.G.)
| | - Gert Folkerts
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584 CG, Utrecht, The Netherlands (M.T.v.D., G.F., J.G., S.B.); and Danone Nutricia Research, 3584 CT, Utrecht, The Netherlands (J.G.)
| | - Johan Garssen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584 CG, Utrecht, The Netherlands (M.T.v.D., G.F., J.G., S.B.); and Danone Nutricia Research, 3584 CT, Utrecht, The Netherlands (J.G.)
| | - Saskia Braber
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584 CG, Utrecht, The Netherlands (M.T.v.D., G.F., J.G., S.B.); and Danone Nutricia Research, 3584 CT, Utrecht, The Netherlands (J.G.)
| |
Collapse
|
17
|
A simplified method for bronchoalveolar lavage in mice by orotracheal intubation avoiding tracheotomy. Biotechniques 2021; 71:534-537. [PMID: 34517774 DOI: 10.2144/btn-2021-0022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Bronchoalveolar lavage (BAL) represents an important method to sample immune cells and soluble substances from the lungs of humans and animals suffering from respiratory disease. The mouse is the most commonly used model organism to study lung disease. Performing BAL in mice is difficult due to their small size and the currently used method requires tracheotomy, a complex and time-consuming procedure. Here, we describe a simple alternative procedure that avoids this step. To perform the BAL, a rigid, olive tip cannula is inserted from the mouth into the trachea under visual inspection. This novel method requires minimal training, is simple, fast, inexpensive and should be useful for researchers studying mouse models of human lung disease.
Collapse
|
18
|
Xie M, Park D, Sica GL, Deng X. Bcl2-induced DNA replication stress promotes lung carcinogenesis in response to space radiation. Carcinogenesis 2021; 41:1565-1575. [PMID: 32157295 DOI: 10.1093/carcin/bgaa021] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 02/18/2020] [Accepted: 03/05/2020] [Indexed: 11/12/2022] Open
Abstract
Space radiation is characterized by high-linear energy transfer (LET) ionizing radiation. The relationships between the early biological effects of space radiation and the probability of cancer in humans are poorly understood. Bcl2 not only functions as a potent antiapoptotic molecule but also as an oncogenic protein that induces DNA replication stress. To test the role and mechanism of Bcl2 in high-LET space radiation-induced lung carcinogenesis, we created lung-targeting Bcl2 transgenic C57BL/6 mice using the CC10 promoter to drive Bcl2 expression selectively in lung tissues. Intriguingly, lung-targeting transgenic Bcl2 inhibits ribonucleotide reductase activity, reduces dNTP pool size and retards DNA replication fork progression in mouse bronchial epithelial cells. After exposure of mice to space radiation derived from 56iron, 28silicon or protons, the incidence of lung cancer was significantly higher in lung-targeting Bcl2 transgenic mice than in wild-type mice, indicating that Bcl2-induced DNA replication stress promotes lung carcinogenesis in response to space radiation. The findings provide some evidence for the relative effectiveness of space radiation and Bcl-2 at inducing lung cancer in mice.
Collapse
Affiliation(s)
- Maohua Xie
- Department of Radiation Oncology, Emory University School of Medicine and Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Dongkyoo Park
- Department of Radiation Oncology, Emory University School of Medicine and Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Gabriel L Sica
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine and Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Xingming Deng
- Department of Radiation Oncology, Emory University School of Medicine and Winship Cancer Institute of Emory University, Atlanta, GA, USA
| |
Collapse
|
19
|
Sena IFG, Rocha BGS, Picoli CC, Santos GSP, Costa AC, Gonçalves BOP, Garcia APV, Soltani-Asl M, Coimbra-Campos LMC, Silva WN, Costa PAC, Pinto MCX, Amorim JH, Azevedo VAC, Resende RR, Heller D, Cassali GD, Mintz A, Birbrair A. C(3)1-TAg in C57BL/6 J background as a model to study mammary tumor development. Histochem Cell Biol 2021; 156:165-182. [PMID: 34003355 DOI: 10.1007/s00418-021-01995-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/09/2021] [Indexed: 02/06/2023]
Abstract
Diagnosis and prognosis of breast cancer is based on disease staging identified through histopathological and molecular biology techniques. Animal models are used to gain mechanistic insights into the development of breast cancer. C(3)1-TAg is a genetically engineered mouse model that develops mammary cancer. However, carcinogenesis caused by this transgene was characterized in the Friend Virus B (FVB) background. As most genetic studies are done in mice with C57BL/6 J background, we aimed to define the histological alterations in C3(1)-TAg C57BL/6 J animals. Our results showed that C3(1)-TAg animals with C57BL/6 J background develop solid-basaloid adenoid cystic carcinomas with increased fibrosis, decreased area of adipocytes, and a high proliferative index, which are triple-negative for progesterone, estrogen, and human epidermal growth factor receptor 2 (HER2) receptors. Our results also revealed that tumor development is slower in the C57BL/6 J background when compared with the FVB strain, providing a better model to study the different stages in breast cancer progression.
Collapse
Affiliation(s)
- Isadora F G Sena
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Beatriz G S Rocha
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Caroline C Picoli
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Gabryella S P Santos
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Alinne C Costa
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Bryan O P Gonçalves
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Ana Paula V Garcia
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Maryam Soltani-Asl
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | | | - Walison N Silva
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Pedro A C Costa
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Mauro C X Pinto
- Laboratory of Neuropharmacology and Neurochemistry, Institute of Biological Sciences, Federal University of Goiás, Goiânia, GO, Brazil
| | - Jaime H Amorim
- Center of Biological Sciences and Health, Federal University of West Bahia, Barreiras, BA, Brazil
| | - Vasco A C Azevedo
- Cellular and Molecular Genetics Laboratory, Department of Genetics, Ecology and Evolution, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Rodrigo R Resende
- Department of Biochemistry and Immunology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Debora Heller
- Hospital Israelita Albert Einstein, São Paulo, Brazil.,Cruzeiro Do Sul University, São Paulo, Brazil
| | - Geovanni D Cassali
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Akiva Mintz
- Department of Radiology, Columbia University Medical Center, New York, NY, USA
| | - Alexander Birbrair
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil. .,Department of Radiology, Columbia University Medical Center, New York, NY, USA.
| |
Collapse
|
20
|
Ma Q, Grigorescu M, Schreiber A, Kettritz R, Lindenmeyer M, Anders HJ, Steiger S. Genetic Background but Not Intestinal Microbiota After Co-Housing Determines Hyperoxaluria-Related Nephrocalcinosis in Common Inbred Mouse Strains. Front Immunol 2021; 12:673423. [PMID: 33968083 PMCID: PMC8100042 DOI: 10.3389/fimmu.2021.673423] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Accepted: 04/06/2021] [Indexed: 12/20/2022] Open
Abstract
Calcium oxalate (CaOx) crystal formation, aggregation and growth is a common cause of kidney stone disease and nephrocalcinosis-related chronic kidney disease (CKD). Genetically modified mouse strains are frequently used as an experimental tool in this context but observed phenotypes may also relate to the genetic background or intestinal microbiota. We hypothesized that the genetic background or intestinal microbiota of mice determine CaOx crystal deposition and thus the outcome of nephrocalcinosis. Indeed, Casp1-/-, Cybb-/- or Casp1-/-/Cybb-/- knockout mice on a 129/C57BL/6J (B6J) background that were fed an oxalate-rich diet for 14 days did neither encounter intrarenal CaOx crystal deposits nor nephrocalcinosis-related CKD. To test our assumption, we fed C57BL/6N (B6N), 129, B6J and Balb/c mice an oxalate-rich diet for 14 days. Only B6N mice displayed CaOx crystal deposits and developed CKD associated with tubular injury, inflammation and interstitial fibrosis. Intrarenal mRNA expression profiling of 64 known nephrocalcinosis-related genes revealed that healthy B6N mice had lower mRNA levels of uromodulin (Umod) compared to the other three strains. Feeding an oxalate-rich diet caused an increase in uromodulin protein expression and CaOx crystal deposition in the kidney as well as in urinary uromodulin excretion in B6N mice but not 129, B6J and Balb/c mice. However, backcrossing 129 mice on a B6N background resulted in a gradual increase in CaOx crystal deposits from F2 to F7, of which all B6N/129 mice from the 7th generation developed CaOx-related nephropathy similar to B6N mice. Co-housing experiments tested for a putative role of the intestinal microbiota but B6N co-housed with 129 mice or B6N/129 (3rd and 6th generation) mice did not affect nephrocalcinosis. In summary, genetic background but not the intestinal microbiome account for strain-specific crystal formation and, the levels of uromodulin secretion may contribute to this phenomenon. Our results imply that only littermate controls of the identical genetic background strain are appropriate when performing knockout mouse studies in this context, while co-housing is optional.
Collapse
Affiliation(s)
- Qiuyue Ma
- Division of Nephrology, Department of Medicine IV, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Melissa Grigorescu
- Division of Nephrology, Department of Medicine IV, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Adrian Schreiber
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Experimental and Clinical Research Center, Charité-Universitätsmedizin Berlin and Max Delbrueck Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Ralph Kettritz
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Experimental and Clinical Research Center, Charité-Universitätsmedizin Berlin and Max Delbrueck Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Maja Lindenmeyer
- III. Department of Medicine University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Hans-Joachim Anders
- Division of Nephrology, Department of Medicine IV, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Stefanie Steiger
- Division of Nephrology, Department of Medicine IV, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| |
Collapse
|
21
|
Sura R, Hutt J, Morgan S. Opinion on the Use of Animal Models in Nonclinical Safety Assessment: Pros and Cons. Toxicol Pathol 2021; 49:990-995. [PMID: 33827334 DOI: 10.1177/01926233211003498] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Nonclinical evaluation of human safety risks for new chemical entities (NCEs) is primarily conducted in conventional healthy animals (CHAs); however, in certain instances, animal models of diseases (AMDs) can play a critical role in the understanding of human health risks. Animal models of diseases may be especially important when there is a need to understand how disease conditions associated with the intended indication might impact risk assessment of NCEs or when CHAs lack the human-specific target of interest (receptor, etc). Although AMDs have potential benefits over CHAs, they also have limitations. Understanding these limitations and optimizing the AMDs of interest should be done prior to proceeding with studies that will guide development of NCE. The purpose of this manuscript is to provide an overview of the major pros and cons of utilization of AMDs in nonclinical safety assessment.
Collapse
Affiliation(s)
| | - Julie Hutt
- Greenfield Pathology Services, Inc., Greenfield, IN, USA
| | | |
Collapse
|
22
|
van Weele LJ, Scheeren FA, Cai S, Kuo AH, Qian D, Ho WHD, Clarke MF. Depletion of Trp53 and Cdkn2a Does Not Promote Self-Renewal in the Mammary Gland but Amplifies Proliferation Induced by TNF-α. Stem Cell Reports 2021; 16:228-236. [PMID: 33482103 PMCID: PMC7878826 DOI: 10.1016/j.stemcr.2020.12.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 12/18/2020] [Accepted: 12/19/2020] [Indexed: 11/08/2022] Open
Abstract
The mammary epithelium undergoes several rounds of extensive proliferation during the female reproductive cycle. Its expansion is a tightly regulated process, fueled by the mammary stem cells and these cells' unique property of self-renewal. Sufficient new cells have to be produced to maintain the integrity of a tissue, but excessive proliferation resulting in tumorigenesis needs to be prevented. Three well-known tumor suppressors, p53, p16INK4a, and p19ARF, have been connected to the limiting of stem cell self-renewal and proliferation. Here we investigate the roles of these three proteins in the regulation of self-renewal and proliferation of mammary epithelial cells. Using mammary epithelial-specific mouse models targeting Trp53 and Cdkn2a, the gene coding for p16INK4a and p19ARF, we demonstrate that p53, p16INK4a, and p19ARF do not play a significant role in the limitation of normal mammary epithelium self-renewal and proliferation, whereas in the presence of the inflammatory cytokine TNF-α, Trp53−/−Cdkn2a−/− mammary basal cells exhibit amplified proliferation. p53, p16INK4a, and p19ARF do not limit self-renewal of mammary epithelial cells p53, p16INK4a, and p19ARF do not limit proliferation of mammary epithelial cells TNF-α stimulates mammary basal cell organoid formation and proliferation Trp53−/−Cdkn2a−/− organoids are sensitized to TNF-α-induced proliferation
Collapse
Affiliation(s)
- Linda J van Weele
- Institute for Stem Cell Biology and Regenerative Medicine, School of Medicine, Stanford University, Stanford, CA, USA
| | - Ferenc A Scheeren
- Institute for Stem Cell Biology and Regenerative Medicine, School of Medicine, Stanford University, Stanford, CA, USA
| | - Shang Cai
- Institute for Stem Cell Biology and Regenerative Medicine, School of Medicine, Stanford University, Stanford, CA, USA
| | - Angera H Kuo
- Institute for Stem Cell Biology and Regenerative Medicine, School of Medicine, Stanford University, Stanford, CA, USA
| | - Dalong Qian
- Institute for Stem Cell Biology and Regenerative Medicine, School of Medicine, Stanford University, Stanford, CA, USA
| | - William H D Ho
- Institute for Stem Cell Biology and Regenerative Medicine, School of Medicine, Stanford University, Stanford, CA, USA; Department of Stem Cell Biotechnology, California State University Channel Islands, Camarillo, CA, USA
| | - Michael F Clarke
- Institute for Stem Cell Biology and Regenerative Medicine, School of Medicine, Stanford University, Stanford, CA, USA.
| |
Collapse
|
23
|
Medert R, Pironet A, Bacmeister L, Segin S, Londoño JEC, Vennekens R, Freichel M. Genetic background influences expression and function of the cation channel TRPM4 in the mouse heart. Basic Res Cardiol 2020; 115:70. [PMID: 33205255 PMCID: PMC7671982 DOI: 10.1007/s00395-020-00831-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 11/02/2020] [Indexed: 01/21/2023]
Abstract
Transient receptor potential melastatin 4 (TRPM4) cation channels act in cardiomyocytes as a negative modulator of the L-type Ca2+ current. Ubiquitous Trpm4 deletion in mice leads to an increased β-adrenergic inotropy in healthy mice as well as after myocardial infarction. In this study, we set out to investigate cardiac inotropy in mice with cardiomyocyte-specific Trpm4 deletion. The results guided us to investigate the relevance of TRPM4 for catecholamine-evoked Ca2+ signaling in cardiomyocytes and inotropy in vivo in TRPM4-deficient mouse models of different genetic background. Cardiac hemodynamics were investigated using pressure-volume analysis. Surprisingly, an increased β-adrenergic inotropy was observed in global TRPM4-deficient mice on a 129SvJ genetic background, but the inotropic response was unaltered in mice with global and cardiomyocyte-specific TRPM4 deletion on the C57Bl/6N background. We found that the expression of TRPM4 proteins is about 78 ± 10% higher in wild-type mice on the 129SvJ versus C57Bl/6N background. In accordance with contractility measurements, our analysis of the intracellular Ca2+ transients revealed an increase in ISO-evoked Ca2+ rise in Trpm4-deficient cardiomyocytes of the 129SvJ strain, but not of the C57Bl/6N strain. No significant differences were observed between the two mouse strains in the expression of other regulators of cardiomyocyte Ca2+ homeostasis. We conclude that the relevance of TRPM4 for cardiac contractility depends on homeostatic TRPM4 expression levels or the genetic endowment in different mouse strains as well as on the health/disease status. Therefore, the concept of inhibiting TRPM4 channels to improve cardiac contractility needs to be carefully explored in specific strains and species and prospectively in different genetically diverse populations of patients.
Collapse
Affiliation(s)
- Rebekka Medert
- Institute of Pharmacology, Heidelberg University, im Neuenheimer Feld 366, 69120, Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site, Heidelberg/Mannheim, Germany
| | - Andy Pironet
- Laboratory of Ion Channel Research, TRP Research Platform Leuven, VIB Center for Brain and Disease Research, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Lucas Bacmeister
- Institute of Pharmacology, Heidelberg University, im Neuenheimer Feld 366, 69120, Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site, Heidelberg/Mannheim, Germany
| | - Sebastian Segin
- Institute of Pharmacology, Heidelberg University, im Neuenheimer Feld 366, 69120, Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site, Heidelberg/Mannheim, Germany
| | - Juan E Camacho Londoño
- Institute of Pharmacology, Heidelberg University, im Neuenheimer Feld 366, 69120, Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site, Heidelberg/Mannheim, Germany
| | - Rudi Vennekens
- Laboratory of Ion Channel Research, TRP Research Platform Leuven, VIB Center for Brain and Disease Research, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Marc Freichel
- Institute of Pharmacology, Heidelberg University, im Neuenheimer Feld 366, 69120, Heidelberg, Germany.
- DZHK (German Centre for Cardiovascular Research), Partner Site, Heidelberg/Mannheim, Germany.
| |
Collapse
|
24
|
Hsieh YL, Su FY, Tsai LK, Huang CC, Ko YL, Su LW, Chen KY, Shih HM, Hu CM, Lee WH. NPGPx-Mediated Adaptation to Oxidative Stress Protects Motor Neurons from Degeneration in Aging by Directly Modulating O-GlcNAcase. Cell Rep 2020; 29:2134-2143.e7. [PMID: 31747588 DOI: 10.1016/j.celrep.2019.10.053] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 07/10/2019] [Accepted: 10/11/2019] [Indexed: 12/14/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS), the most common motor neuron disease, usually occurs in middle-aged people. However, the molecular basis of age-related cumulative stress in ALS pathogenesis remains elusive. Here, we found that mice deficient in NPGPx (GPx7), an oxidative stress sensor, develop ALS-like phenotypes, including paralysis, muscle denervation, and motor neurons loss. Unlike normal spinal motor neurons that exhibit elevated O-GlcNAcylation against age-dependent oxidative stress, NPGPx-deficient spinal motor neurons fail to boost O-GlcNAcylation and exacerbate ROS accumulation, leading to cell death. Mechanistically, stress-activated NPGPx inhibits O-GlcNAcase (OGA) through disulfide bonding to fine-tune global O-GlcNAcylation. Pharmacological inhibition of OGA rescues spinal motor neuron loss in aged NPGPx-deficient mice. Furthermore, expression of NPGPx in ALS patients is significantly lower than in unaffected adults. These results suggest that NPGPx modulates O-GlcNAcylation by inhibiting OGA to cope with age-dependent oxidative stress and protect motor neurons from degeneration, providing a potential therapeutic axis for ALS.
Collapse
Affiliation(s)
- Yung-Lin Hsieh
- Genomics Research Center, Academia Sinica, Taipei 115, Taiwan
| | - Fang-Yi Su
- Genomics Research Center, Academia Sinica, Taipei 115, Taiwan; Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei 112, Taiwan
| | - Li-Kai Tsai
- Department of Neurology, National Taiwan University Hospital, Taipei 100, Taiwan
| | | | - Yi-Ling Ko
- Genomics Research Center, Academia Sinica, Taipei 115, Taiwan
| | - Li-Wen Su
- Genomics Research Center, Academia Sinica, Taipei 115, Taiwan
| | - Kai-Yun Chen
- Graduate Institute of Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan
| | - Hsiu-Ming Shih
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan; Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli 350, Taiwan
| | - Chun-Mei Hu
- Genomics Research Center, Academia Sinica, Taipei 115, Taiwan
| | - Wen-Hwa Lee
- Genomics Research Center, Academia Sinica, Taipei 115, Taiwan; Department of Biological Chemistry, University of California, Irvine, Irvine, CA 92697, USA; Drug Development Center, China Medical University, Taichung 404, Taiwan.
| |
Collapse
|
25
|
Andrews RN, Bloomer EG, Olson JD, Hanbury DB, Dugan GO, Whitlow CT, Cline JM. Non-Human Primates Receiving High-Dose Total-Body Irradiation are at Risk of Developing Cerebrovascular Injury Years Postirradiation. Radiat Res 2020; 194:277-287. [PMID: 32942304 PMCID: PMC7583660 DOI: 10.1667/rade-20-00051.1] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 06/08/2020] [Indexed: 12/15/2022]
Abstract
Nuclear accidents and acts of terrorism have the potential to expose thousands of people to high-dose total-body iradiation (TBI). Those who survive the acute radiation syndrome are at risk of developing chronic, degenerative radiation-induced injuries [delayed effects of acute radiation (DEARE)] that may negatively affect quality of life. A growing body of literature suggests that the brain may be vulnerable to radiation injury at survivable doses, yet the long-term consequences of high-dose TBI on the adult brain are unclear. Herein we report the occurrence of lesions consistent with cerebrovascular injury, detected by susceptibility-weighted magnetic resonance imaging (MRI), in a cohort of non-human primate [(NHP); rhesus macaque, Macaca mulatta] long-term survivors of high-dose TBI (1.1-8.5 Gy). Animals were monitored longitudinally with brain MRI (approximately once every three years). Susceptibility-weighted images (SWI) were reviewed for hypointensities (cerebral microbleeds and/or focal necrosis). SWI hypointensities were noted in 13% of irradiated NHP; lesions were not observed in control animals. A prior history of exposure was correlated with an increased risk of developing a lesion detectable by MRI (P = 0.003). Twelve of 16 animals had at least one brain lesion present at the time of the first MRI evaluation; a subset of animals (n = 7) developed new lesions during the surveillance period (3.7-11.3 years postirradiation). Lesions occurred with a predilection for white matter and the gray-white matter junction. The majority of animals with lesions had one to three SWI hypointensities, but some animals had multifocal disease (n = 2). Histopathologic evaluation of deceased animals within the cohort (n = 3) revealed malformation of the cerebral vasculature and remodeling of the blood vessel walls. There was no association between comorbid diabetes mellitus or hypertension with SWI lesion status. These data suggest that long-term TBI survivors may be at risk of developing cerebrovascular injury years after irradiation.
Collapse
Affiliation(s)
- Rachel N. Andrews
- Department of Radiation Oncology, Section of Radiation Biology, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina 27157
- Department of Pathology, Section on Comparative Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina 27157
- Department of Wake Forest Baptist Comprehensive Cancer Center, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina 27157
| | - Ethan G. Bloomer
- University of Florida, College of Veterinary Medicine, Gainesville, Florida 32608
| | - John D. Olson
- Department of Pathology, Section on Comparative Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina 27157
| | - David B. Hanbury
- Department of Psychology, Averett University, Danville, Virginia 24541
| | - Gregory O. Dugan
- Department of Pathology, Section on Comparative Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina 27157
| | - Christopher T. Whitlow
- Department of Wake Forest Baptist Comprehensive Cancer Center, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina 27157
- Department of Radiology, Section of Neuroradiology, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina 27157
- Department of Biomedical Engineering, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina 27157
- Department of Biostatistics and Data Science, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina 27157
| | - J. Mark Cline
- Department of Pathology, Section on Comparative Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina 27157
| |
Collapse
|
26
|
Enriquez J, Mims BMD, Trasti S, Furr KL, Grisham MB. Genomic, microbial and environmental standardization in animal experimentation limiting immunological discovery. BMC Immunol 2020; 21:50. [PMID: 32878597 PMCID: PMC7464063 DOI: 10.1186/s12865-020-00380-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 08/25/2020] [Indexed: 02/07/2023] Open
Abstract
Background The use of inbred mice housed under standardized environmental conditions has been critical in identifying immuno-pathological mechanisms in different infectious and inflammatory diseases as well as revealing new therapeutic targets for clinical trials. Unfortunately, only a small percentage of preclinical intervention studies using well-defined mouse models of disease have progressed to clinically-effective treatments in patients. The reasons for this lack of bench-to-bedside transition are not completely understood; however, emerging data suggest that genetic diversity and housing environment may greatly influence muring immunity and inflammation. Results Accumulating evidence suggests that certain immune responses and/or disease phenotypes observed in inbred mice may be quite different than those observed in their outbred counterparts. These differences have been thought to contribute to differing immune responses to foreign and/or auto-antigens in mice vs. humans. There is also a growing literature demonstrating that mice housed under specific pathogen free conditions possess an immature immune system that remarkably affects their ability to respond to pathogens and/or inflammation when compared with mice exposed to a more diverse spectrum of microorganisms. Furthermore, recent studies demonstrate that mice develop chronic cold stress when housed at standard animal care facility temperatures (i.e. 22–24 °C). These temperatures have been shown alter immune responses to foreign and auto-antigens when compared with mice housed at their thermo-neutral body temperature of 30–32 °C. Conclusions Exposure of genetically diverse mice to a spectrum of environmentally-relevant microorganisms at housing temperatures that approximate their thermo-neutral zone may improve the chances of identifying new and more potent therapeutics to treat infectious and inflammatory diseases.
Collapse
Affiliation(s)
- Josue Enriquez
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, 3601 4th Street STOP 6591, Lubbock, TX, 79430-6591, USA
| | - Brianyell Mc Daniel Mims
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, 3601 4th Street STOP 6591, Lubbock, TX, 79430-6591, USA
| | - Scott Trasti
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, 3601 4th Street STOP 6591, Lubbock, TX, 79430-6591, USA.,Laboratory Animal Research Center, Texas Tech University Health Sciences Center, Lubbock, TX, 79430, USA
| | - Kathryn L Furr
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, 3601 4th Street STOP 6591, Lubbock, TX, 79430-6591, USA
| | - Matthew B Grisham
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, 3601 4th Street STOP 6591, Lubbock, TX, 79430-6591, USA.
| |
Collapse
|
27
|
Martin MD, Badovinac VP, Griffith TS. CD4 T Cell Responses and the Sepsis-Induced Immunoparalysis State. Front Immunol 2020; 11:1364. [PMID: 32733454 PMCID: PMC7358556 DOI: 10.3389/fimmu.2020.01364] [Citation(s) in RCA: 106] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 05/28/2020] [Indexed: 12/20/2022] Open
Abstract
Sepsis remains a major cause of death in the United States and worldwide, and costs associated with treating septic patients place a large burden on the healthcare industry. Patients who survive the acute phase of sepsis display long-term impairments in immune function due to reductions in numbers and function of many immune cell populations. This state of chronic immunoparalysis renders sepsis survivors increasingly susceptible to infection with newly or previously encountered infections. CD4 T cells play important roles in the development of cellular and humoral immune responses following infection. Understanding how sepsis impacts the CD4 T cell compartment is critical for informing efforts to develop treatments intended to restore immune system homeostasis following sepsis. This review will focus on the current understanding of how sepsis impacts the CD4 T cell responses, including numerical representation, repertoire diversity, phenotype and effector functionality, subset representation (e.g., Th1 and Treg frequency), and therapeutic efforts to restore CD4 T cell numbers and function following sepsis. Additionally, we will discuss recent efforts to model the acute sepsis phase and resulting immune dysfunction using mice that have previously encountered infection, which more accurately reflects the immune system of humans with a history of repeated infection throughout life. A thorough understanding of how sepsis impacts CD4 T cells based on previous studies and new models that accurately reflect the human immune system may improve translational value of research aimed at restoring CD4 T cell-mediated immunity, and overall immune fitness following sepsis.
Collapse
Affiliation(s)
- Matthew D. Martin
- Department of Urology, University of Minnesota, Minneapolis, MN, United States
| | - Vladimir P. Badovinac
- Department of Pathology, University of Iowa, Iowa City, IA, United States
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA, United States
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, United States
| | - Thomas S. Griffith
- Department of Urology, University of Minnesota, Minneapolis, MN, United States
- Microbiology, Immunology, and Cancer Biology PhD Program, University of Minnesota, Minneapolis, MN, United States
- Center for Immunology, University of Minnesota, Minneapolis, MN, United States
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, United States
- Minneapolis VA Healthcare System, Minneapolis, MN, United States
| |
Collapse
|
28
|
Richter AM, Woods ML, Küster MM, Walesch SK, Braun T, Boettger T, Dammann RH. RASSF10 is frequently epigenetically inactivated in kidney cancer and its knockout promotes neoplasia in cancer prone mice. Oncogene 2020; 39:3114-3127. [PMID: 32047266 PMCID: PMC7142015 DOI: 10.1038/s41388-020-1195-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 01/21/2020] [Accepted: 01/27/2020] [Indexed: 12/22/2022]
Abstract
Kidney cancer incidences are rising globally, thereby fueling the demand for targeted therapies and precision medicine. In our previous work, we have identified and characterized the Ras-Association Domain Family encoding ten members that are often aberrantly expressed in human cancers. In this study, we created and analyzed the Rassf10 knockout mice. Here we show that Rassf10 haploinsufficiency promotes neoplasia formation in two established mouse cancer models (Rassf1A-/- and p53-/-). Haploinsufficient Rassf10 knockout mice were significantly prone to various diseases including lymphoma (Rassf1A-/- background) and thymoma (p53-/- background). Especially Rassf10-/- and p53-deficient mice exhibited threefold increased rates of kidney cysts compared with p53-/- controls. Moreover, we observed that in human kidney cancer, RASSF10 is frequently epigenetically inactivated by its CpG island promoter hypermethylation. Primary tumors of renal clear cell and papillary cell carcinoma confirmed that RASSF10 methylation is associated with decreased expression in comparison to normal kidney tissue. In independent data sets, we could validate that RASSF10 inactivation clinically correlated with decreased survival and with progressed disease state of kidney cancer patients and polycystic kidney size. Functionally, we revealed that the loss of Rassf10 was significantly associated with upregulation of KRAS signaling and MYC expression. In summary, we could show that Rassf10 functions as a haploinsufficient tumor suppressor. In combination with other markers, RASSF10 silencing can serve as diagnostic and prognostic cancer biomarker in kidney diseases.
Collapse
Affiliation(s)
- Antje M Richter
- Institute for Genetics, University of Giessen, 35392, Giessen, Germany. .,Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany.
| | - Michelle L Woods
- Institute for Genetics, University of Giessen, 35392, Giessen, Germany
| | - Miriam M Küster
- Institute for Genetics, University of Giessen, 35392, Giessen, Germany
| | - Sara K Walesch
- Institute for Genetics, University of Giessen, 35392, Giessen, Germany
| | - Thomas Braun
- Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany.,German Center for Lung Research (DZL), Universities of Giessen and Marburg Lung Center, 35392, Giessen, Germany
| | - Thomas Boettger
- Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Reinhard H Dammann
- Institute for Genetics, University of Giessen, 35392, Giessen, Germany. .,German Center for Lung Research (DZL), Universities of Giessen and Marburg Lung Center, 35392, Giessen, Germany.
| |
Collapse
|
29
|
Hashimoto R, Lanier GM, Dhagia V, Joshi SR, Jordan A, Waddell I, Tuder R, Stenmark KR, Wolin MS, McMurtry IF, Gupte SA. Pluripotent hematopoietic stem cells augment α-adrenergic receptor-mediated contraction of pulmonary artery and contribute to the pathogenesis of pulmonary hypertension. Am J Physiol Lung Cell Mol Physiol 2020; 318:L386-L401. [PMID: 31913656 PMCID: PMC7052680 DOI: 10.1152/ajplung.00327.2019] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 12/10/2019] [Accepted: 12/19/2019] [Indexed: 12/13/2022] Open
Abstract
Pulmonary hypertension (PH) is a multicellular and progressive disease with a high mortality rate. Among many cell types, hematopoietic stem cells (HSCs) are incriminated in the pathogenesis of PH. However, our understanding of the mechanisms that increase HSCs in blood and lungs of hypertensive animals or patients and the role played by HSCs in the pathogenesis of PH remains elusive. Studies suggest that glycolysis is critical for the survival and growth of HSCs. In various cell types from hypertensive lungs of animals and patients, glycolysis and the glucose-6-phosphate dehydrogenase (G6PD) activity are increased. Herein, we demonstrated in mice that chronic hypoxia increased HSCs (CD34+, CD117+, CD133+, CD34+/CD117+, and CD34+/CD133+) in bone marrow and blood and around hypertensive pulmonary arteries in a time-dependent manner. Intriguingly, we found fewer CD133+ cells in the bone marrow of C57BL/6 mice compared with Sv129J mice, and C57BL mice developed less severe chronic hypoxia-elicited PH and heart failure than Sv129J mice. Similarly, the numbers of CD34+ and CD117+ cells in blood of patients with pulmonary arterial hypertension (PAH) were higher (>3-fold) compared with healthy individuals. By allogeneic bone marrow transplantation, we found that GFP+ bone marrow cells infiltrated the lungs and accumulated around the pulmonary arteries in lungs of hypoxic mice, and these cells contributed to increased α-adrenergic receptor-mediated contraction of the pulmonary artery cultured in hypoxia. Inhibition of G6PD activity with (3β,5α)-3,21-dihydroxypregnan-20-one, a novel and potent G6PD inhibitor, decreased HSCs in bone marrow, blood, and lungs of hypoxic mice and reduced α-agonist-induced contraction of the pulmonary artery and established hypoxia-induced PH. We did not observe CD133+ cells around the pulmonary arteries in the lungs of chronically hypoxic G6PD-deficient mice. Furthermore, knockdown of G6PD and inhibition of G6PD activity: 1) downregulated canonical and noncanonical Wnt and Fzd receptors genes; 2) upregulated Bmpr1a; 3) decreased Cxcl12, and 4) reduced HSC (CD117+ and CD133+) numbers. In all, our findings demonstrate unexpected function for bone marrow-derived HSCs in augmenting α-adrenergic receptor-mediated contraction of pulmonary arteries and remodeling of pulmonary arteries that contribute to increase pulmonary vascular resistance in PAH patients and hypoxic mice and suggest that G6PD, by regulating expression of genes in the WNT and BMPR signaling, contributed to increase and release of HSCs from the bone marrow in response to hypoxic stimuli.
Collapse
Affiliation(s)
- Ryota Hashimoto
- Department of Pharmacology, New York Medical College, Valhalla, New York
| | - Gregg M Lanier
- Department of Cardiology, and Heart and Vascular Institute, Westchester Medical Center and New York Medical College, Valhalla, New York
| | - Vidhi Dhagia
- Department of Pharmacology, New York Medical College, Valhalla, New York
| | - Sachindra R Joshi
- Department of Pharmacology, New York Medical College, Valhalla, New York
| | - Allan Jordan
- Drug Discovery Unit, Cancer Research, UK Manchester Institute, University of Manchester, Manchester, United Kingdom
| | - Ian Waddell
- Drug Discovery Unit, Cancer Research, UK Manchester Institute, University of Manchester, Manchester, United Kingdom
| | - Rubin Tuder
- Department of Pathology, University of Colorado Health Center, Denver, Colorado
| | - Kurt R Stenmark
- Department of Pediatrics, University of Colorado Health Center, Denver, Colorado
| | - Michael S Wolin
- Department of Physiology, New York Medical College, Valhalla, New York
| | - Ivan F McMurtry
- Department of Pharmacology and Medicine, University of South Alabama, Mobile, Alabama
| | - Sachin A Gupte
- Department of Pharmacology, New York Medical College, Valhalla, New York
| |
Collapse
|
30
|
Olivera A, Rivera J. Paradigm Shifts in Mast Cell and Basophil Biology and Function: An Emerging View of Immune Regulation in Health and Disease. Methods Mol Biol 2020; 2163:3-31. [PMID: 32766962 DOI: 10.1007/978-1-0716-0696-4_1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
The physiological role of the mast cell and basophil has for many years remained enigmatic. In this chapter, we briefly summarize some of the more recent studies that shed new light on the role of mast cells and basophils in health and disease. What we gain from these studies is a new appreciation for mast cells and basophils as sentinels in host defense and a further understanding that dysregulation of mast cell and basophil function can be a component of various diseases other than allergies. Perhaps the most important insight reaped from this work is the increasing awareness that mast cells and basophils can function as immunoregulatory cells that modulate the immune response in health and disease. Collectively, the recent knowledge provides new challenges and opportunities toward the development of novel therapeutic strategies to augment host protection and modify disease through manipulation of mast cell and basophil function.
Collapse
Affiliation(s)
- Ana Olivera
- Molecular Immunology Section, Laboratory of Molecular Immunogenetics, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA.
- Mast Cell Biology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health (NIH), Bethesda, MD, USA.
| | - Juan Rivera
- Molecular Immunology Section, Laboratory of Molecular Immunogenetics, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
31
|
Radaelli E, Santagostino SF, Sellers RS, Brayton CF. Immune Relevant and Immune Deficient Mice: Options and Opportunities in Translational Research. ILAR J 2019; 59:211-246. [PMID: 31197363 PMCID: PMC7114723 DOI: 10.1093/ilar/ily026] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 12/03/2018] [Indexed: 12/29/2022] Open
Abstract
In 1989 ILAR published a list and description of immunodeficient rodents used in research. Since then, advances in understanding of molecular mechanisms; recognition of genetic, epigenetic microbial, and other influences on immunity; and capabilities in manipulating genomes and microbiomes have increased options and opportunities for selecting mice and designing studies to answer important mechanistic and therapeutic questions. Despite numerous scientific breakthroughs that have benefitted from research in mice, there is debate about the relevance and predictive or translational value of research in mice. Reproducibility of results obtained from mice and other research models also is a well-publicized concern. This review summarizes resources to inform the selection and use of immune relevant mouse strains and stocks, aiming to improve the utility, validity, and reproducibility of research in mice. Immune sufficient genetic variations, immune relevant spontaneous mutations, immunodeficient and autoimmune phenotypes, and selected induced conditions are emphasized.
Collapse
Affiliation(s)
- Enrico Radaelli
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Sara F Santagostino
- Department of Safety Assessment, Genentech, Inc., South San Francisco, California
| | | | - Cory F Brayton
- Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
32
|
Adelöf J, Ross JM, Lazic SE, Zetterberg M, Wiseman J, Hernebring M. Conclusions from a behavioral aging study on male and female F2 hybrid mice on age-related behavior, buoyancy in water-based tests, and an ethical method to assess lifespan. Aging (Albany NY) 2019; 11:7150-7168. [PMID: 31509518 PMCID: PMC6756906 DOI: 10.18632/aging.102242] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 08/22/2019] [Indexed: 12/21/2022]
Abstract
Due to strain-specific behavioral idiosyncrasies, inbred mouse strains are suboptimal research models for behavioral aging studies. The aim of this study is to determine age-related behavioral changes of F2 hybrid C57BL/6NxBALB/c male and female mice. Lifespan was followed (nmales=48, nfemales=51) and cohorts of mature adult (7 months), middle-aged (15 months), and old mice (22 months of age; n=7-12 per group) were assessed regarding open-field activity, exploration, passive avoidance learning/memory, and depressive-like behavior. We found that both males and females demonstrated decreased exploratory behavior with age, while memory and depressive-like behavior were maintained. Females exhibited enhanced depressive-like behavior compared to males; however, a correlation between fat mass and swimming activity in the test directly accounted for 30-46% of this behavioral sex difference. In addition, we suggest a method to qualitatively estimate natural lifespan from survival analyses in which animals with signs of pain or severe disease are euthanized. This is, to our knowledge, the first behavioral study to consider both sex and aging in hybrid mice. We here define decreased exploratory behavior as a conserved hallmark of aging independent of sex, highlight the effect of buoyancy in water tests, and provide a method to assay lifespan with reduced animal suffering.
Collapse
Affiliation(s)
- Julia Adelöf
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg 41390, Sweden.,Discovery Biology, Discovery Sciences, R&D AstraZeneca, Gothenburg, Mölndal 43153, Sweden
| | - Jaime M Ross
- Department of Genetics, Paul F. Glenn Center for the Biology of Aging, Harvard Medical School, Boston, MA 02215, USA.,Department of Neuroscience, Biomedicum, Karolinska Institutet, Stockholm 17165, Sweden
| | - Stanley E Lazic
- Quantitative Biology, Discovery Sciences, R&D AstraZeneca, Cambridge CB4 0WG, UK.,Current address: Prioris.ai Inc., Ottawa K2P 2N2, Canada
| | - Madeleine Zetterberg
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg 41390, Sweden
| | - John Wiseman
- Discovery Biology, Discovery Sciences, R&D AstraZeneca, Gothenburg, Mölndal 43153, Sweden
| | - Malin Hernebring
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg 41390, Sweden.,Discovery Biology, Discovery Sciences, R&D AstraZeneca, Gothenburg, Mölndal 43153, Sweden
| |
Collapse
|
33
|
Vignier N, Mougenot N, Bonne G, Muchir A. Effect of genetic background on the cardiac phenotype in a mouse model of Emery-Dreifuss muscular dystrophy. Biochem Biophys Rep 2019; 19:100664. [PMID: 31341969 PMCID: PMC6630059 DOI: 10.1016/j.bbrep.2019.100664] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 06/06/2019] [Accepted: 07/02/2019] [Indexed: 01/09/2023] Open
Abstract
A-type lamins gene (LMNA) mutations cause an autosomal dominant inherited form of Emery-Dreifuss muscular dystrophy (EDMD). EDMD is characterized by slowly progressive muscle weakness and wasting and dilated cardiomyopathy, often leading to heart failure-related disability. EDMD is highly penetrant with poor prognosis and there is currently no specific therapy available. Clinical variability ranges from early onset with severe presentation in childhood to late onset with slow progression in adulthood. Genetic background is a well-known factor that significantly affects phenotype in several mouse models of human diseases. This phenotypic variability is attributed, at least in part, to genetic modifiers that regulate the disease process. To characterize the phenotype of A-type lamins mutation on different genetic background, we created and phenotyped C57BL/6JRj-LmnaH222P/H222P mice (C57Lmnap.H222P) and compared them with the 129S2/SvPasCrl-LmnaH222P/H222P mice (129Lmnap.H222P). These mouse strains were compared with their respective control strains at multiple time points between 3 and 10 months of age. Both contractile and electrical cardiac muscle functions, as well as survival were characterized. We found that 129Lmnap.H222P mice showed significantly reduced body weight and reduced cardiac function earlier than in the C57Lmnap.H222P mice. We also revealed that only 129Lmnap.H222P mice developed heart arrhythmias. The 129Lmnap.H222P model with an earlier onset and more pronounced cardiac phenotype may be more useful for evaluating therapies that target cardiac muscle function, and heart arrhythmias. Mouse model of Emery-Dreifuss muscular dystrophy generated on 129S2/svPasCrl genetic background have a greater life expectancy. Mouse model of Emery-Dreifuss muscular dystrophy generated on 129S2/svPasCrl genetic background showed exacerbated arrhythmia susceptibility. Mouse model of Emery-Dreifuss muscular dystrophy generated on 129S2/svPasCrl genetic background showed more pronounced dilated cardiomyopathy.
Collapse
Affiliation(s)
- Nicolas Vignier
- Sorbonne Université, INSERM UMRS974 Centre de Recherche en Myologie, Institut de Myologie, G.H. Pitié Salpêtrière, F-75651, Paris Cedex 13, France
| | - Nathalie Mougenot
- Sorbonne Université, INSERM UMS28 Phénotypage du petit animal, Faculté de Médecine Pierre et Marie Curie, F-75013, Paris, France
| | - Gisèle Bonne
- Sorbonne Université, INSERM UMRS974 Centre de Recherche en Myologie, Institut de Myologie, G.H. Pitié Salpêtrière, F-75651, Paris Cedex 13, France
| | - Antoine Muchir
- Sorbonne Université, INSERM UMRS974 Centre de Recherche en Myologie, Institut de Myologie, G.H. Pitié Salpêtrière, F-75651, Paris Cedex 13, France
| |
Collapse
|
34
|
Abstract
Sleep deprivation has been reported to be a contributing factor for the epidemic of obesity. However, it is still largely unknown how sleep deprivation contributes to obesity at the transcriptional level. Here, we identified the significantly changed genes and pathways that may contribute to the sleep deprivation-induced obesity by analyzing two online datasets, including mouse obesity database and mouse sleep deprivation database. 298 differentially expressed genes (DEGs) were identified in high fat diet mice as compared to normal diet mice, while 541 DEGs were identified in mice with sleep deprivation when compared with mice with normal sleep. There are 12 common DEGs, such as Saa3 and Plin4, in both comparisons. And six of common DEGs were validated in other Gene Expression Omnibus (GEO) dataset. GO and KEGG pathway analyses revealed 19 common altered pathways, and most of them were metabolic processes, including steroid metabolic process, small molecule metabolic process and cholesterol metabolic process. Notably, we found that Aldoc, Cyp2b10, Nsdhl, Pcsk9, Saa3, Plin4 and Acss2 were involved in most of those altered pathways. Taken together, our study suggests that Saa3, Plin4, Aldoc, Cyp2b10, Nsdhl, Pcsk9 and Acss2 might be involved in sleep deprivation-induced obesity by regulating metabolic processes.
Collapse
Affiliation(s)
- YI WEI
- Nanjing Forest Police College, Nanjing 210023, P. R. China
| |
Collapse
|
35
|
Onos KD, Uyar A, Keezer KJ, Jackson HM, Preuss C, Acklin CJ, O’Rourke R, Buchanan R, Cossette TL, Sukoff Rizzo SJ, Soto I, Carter GW, Howell GR. Enhancing face validity of mouse models of Alzheimer's disease with natural genetic variation. PLoS Genet 2019; 15:e1008155. [PMID: 31150388 PMCID: PMC6576791 DOI: 10.1371/journal.pgen.1008155] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 06/17/2019] [Accepted: 04/24/2019] [Indexed: 01/01/2023] Open
Abstract
Classical laboratory strains show limited genetic diversity and do not harness natural genetic variation. Mouse models relevant to Alzheimer's disease (AD) have largely been developed using these classical laboratory strains, such as C57BL/6J (B6), and this has likely contributed to the failure of translation of findings from mice to the clinic. Therefore, here we test the potential for natural genetic variation to enhance the translatability of AD mouse models. Two widely used AD-relevant transgenes, APPswe and PS1de9 (APP/PS1), were backcrossed from B6 to three wild-derived strains CAST/EiJ, WSB/EiJ, PWK/PhJ, representative of three Mus musculus subspecies. These new AD strains were characterized using metabolic, functional, neuropathological and transcriptional assays. Strain-, sex- and genotype-specific differences were observed in cognitive ability, neurodegeneration, plaque load, cerebrovascular health and cerebral amyloid angiopathy. Analyses of brain transcriptional data showed strain was the greatest driver of variation. We identified significant variation in myeloid cell numbers in wild type mice of different strains as well as significant differences in plaque-associated myeloid responses in APP/PS1 mice between the strains. Collectively, these data support the use of wild-derived strains to better model the complexity of human AD.
Collapse
Affiliation(s)
- Kristen D. Onos
- The Jackson Laboratory, Bar Harbor, Maine, United States of America
| | - Asli Uyar
- The Jackson Laboratory, Bar Harbor, Maine, United States of America
| | - Kelly J. Keezer
- The Jackson Laboratory, Bar Harbor, Maine, United States of America
| | | | - Christoph Preuss
- The Jackson Laboratory, Bar Harbor, Maine, United States of America
| | - Casey J. Acklin
- The Jackson Laboratory, Bar Harbor, Maine, United States of America
| | - Rita O’Rourke
- The Jackson Laboratory, Bar Harbor, Maine, United States of America
| | - Rebecca Buchanan
- The Jackson Laboratory, Bar Harbor, Maine, United States of America
| | | | | | - Ileana Soto
- Department of Biomedical and Translational Sciences, Rowan University, Glassboro, New Jersey, United States of America
| | - Gregory W. Carter
- The Jackson Laboratory, Bar Harbor, Maine, United States of America
- Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, Massachusetts, United States of America
- Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, Maine, United States of America
| | - Gareth R. Howell
- The Jackson Laboratory, Bar Harbor, Maine, United States of America
- Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, Massachusetts, United States of America
- Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, Maine, United States of America
| |
Collapse
|
36
|
Åhlgren J, Voikar V. Experiments done in Black-6 mice: what does it mean? Lab Anim (NY) 2019; 48:171-180. [DOI: 10.1038/s41684-019-0288-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 03/19/2019] [Indexed: 02/06/2023]
|
37
|
Hiraga T, Ninomiya T. Establishment and characterization of a C57BL/6 mouse model of bone metastasis of breast cancer. J Bone Miner Metab 2019; 37:235-242. [PMID: 29667006 DOI: 10.1007/s00774-018-0927-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 04/02/2018] [Indexed: 01/16/2023]
Abstract
Bone is one of the most common sites of metastasis in patients with advanced breast cancer; however, the mechanisms of bone metastasis remain to be fully elucidated. Animal models are essential research tools for investigating the mechanisms of diseases and drug actions. To date, there have only been a few reports in which C57BL/6 mice were used for the study of bone metastases of breast cancer. In the current study, we found that intracardiac inoculation of C57BL/6 mouse-derived parental E0771 breast cancer cells (E0771/Pa) frequently lead to bone metastases in C57BL/6 mice within 2 weeks. The bone-metastatic clone of E0771 (E0771/Bone) established by sequential in vivo selection demonstrated a higher bone-metastatic potential. Although there were no apparent differences in cell morphology or proliferation in monolayer cultures, E0771/Bone showed increased tumorsphere formation in suspension cultures and tumor formation in the orthotopic mammary fat pad in C57BL/6 mice compared with E0771/Pa. Furthermore, E0771/Bone expressed breast cancer stem-like cell surface markers CD24-/CD44+. These findings suggest that E0771/Bone possesses cancer stem-like properties. Quantitative PCR analysis revealed that mRNA expression of parathyroid hormone-related protein (PTHrP), the most common mediator of osteolytic bone metastases of breast cancer, was significantly upregulated in E0771/Bone. Thus, cancer stem-like properties and elevated PTHrP expression likely contribute to the enhanced metastatic potential of E0771/Bone. We believe that this new mouse model is a useful tool for in vivo studies of bone metastases of breast cancer, especially for those using genetically engineered mice with a C57BL/6 background.
Collapse
Affiliation(s)
- Toru Hiraga
- Department of Histology and Cell Biology, Matsumoto Dental University, 1780 Gobara-Hirooka, Shiojiri, Nagano, 399-0781, Japan.
| | - Tadashi Ninomiya
- Division of Hard Tissue Research, Institute for Oral Science, Matsumoto Dental University, Shiojiri, Nagano, Japan
- Department of Anatomy, Nihon University School of Dentistry, Chiyoda-ku, Tokyo, Japan
| |
Collapse
|
38
|
Farkas C, Fuentes-Villalobos F, Rebolledo-Jaramillo B, Benavides F, Castro AF, Pincheira R. Streamlined computational pipeline for genetic background characterization of genetically engineered mice based on next generation sequencing data. BMC Genomics 2019; 20:131. [PMID: 30755158 PMCID: PMC6373082 DOI: 10.1186/s12864-019-5504-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 01/31/2019] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Genetically engineered mice (GEM) are essential tools for understanding gene function and disease modeling. Historically, gene targeting was first done in embryonic stem cells (ESCs) derived from the 129 family of inbred strains, leading to a mixed background or congenic mice when crossed with C57BL/6 mice. Depending on the number of backcrosses and breeding strategies, genomic segments from 129-derived ESCs can be introgressed into the C57BL/6 genome, establishing a unique genetic makeup that needs characterization in order to obtain valid conclusions from experiments using GEM lines. Currently, SNP genotyping is used to detect the extent of 129-derived ESC genome introgression into C57BL/6 recipients; however, it fails to detect novel/rare variants. RESULTS Here, we present a computational pipeline implemented in the Galaxy platform and in BASH/R script to determine genetic introgression of GEM using next generation sequencing data (NGS), such as whole genome sequencing (WGS), whole exome sequencing (WES) and RNA-Seq. The pipeline includes strategies to uncover variants linked to a targeted locus, genome-wide variant visualization, and the identification of potential modifier genes. Although these methods apply to congenic mice, they can also be used to describe variants fixed by genetic drift. As a proof of principle, we analyzed publicly available RNA-Seq data from five congenic knockout (KO) lines and our own RNA-Seq data from the Sall2 KO line. Additionally, we performed target validation using several genetics approaches. CONCLUSIONS We revealed the impact of the 129-derived ESC genome introgression on gene expression, predicted potential modifier genes, and identified potential phenotypic interference in KO lines. Our results demonstrate that our new approach is an effective method to determine genetic introgression of GEM.
Collapse
Affiliation(s)
- C Farkas
- Laboratorio de Transducción de Señales y Cáncer. Departamento de Bioquímica y Biología Molecular. Facultad Cs. Biológicas, Universidad de Concepción, Concepción, Chile
| | - F Fuentes-Villalobos
- Laboratorio de Transducción de Señales y Cáncer. Departamento de Bioquímica y Biología Molecular. Facultad Cs. Biológicas, Universidad de Concepción, Concepción, Chile
| | | | - F Benavides
- Department of Epigenetics and Molecular Carcinogenesis, M.D. Anderson Cancer Center, Smithville, TX, USA
| | - A F Castro
- Laboratorio de Transducción de Señales y Cáncer. Departamento de Bioquímica y Biología Molecular. Facultad Cs. Biológicas, Universidad de Concepción, Concepción, Chile
| | - R Pincheira
- Laboratorio de Transducción de Señales y Cáncer. Departamento de Bioquímica y Biología Molecular. Facultad Cs. Biológicas, Universidad de Concepción, Concepción, Chile.
| |
Collapse
|
39
|
MITCHELL CASSIEM, DAVY BRENDAM, HULVER MATTHEWW, NEILSON ANDREWP, BENNETT BRIANJ, DAVY KEVINP. Does Exercise Alter Gut Microbial Composition? A Systematic Review. Med Sci Sports Exerc 2019; 51:160-167. [DOI: 10.1249/mss.0000000000001760] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
40
|
Characterization of immune cell subtypes in three commonly used mouse strains reveals gender and strain-specific variations. J Transl Med 2019; 99:93-106. [PMID: 30353130 PMCID: PMC6524955 DOI: 10.1038/s41374-018-0137-1] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 08/22/2018] [Accepted: 09/12/2018] [Indexed: 12/21/2022] Open
Abstract
The lack of consensus on bone marrow (BM) and splenic immune cell profiles in preclinical mouse strains complicates comparative analysis across different studies. Although studies have documented relative distribution of immune cells from peripheral blood in mice, similar studies for BM and spleen from naïve mice are lacking. In an effort to establish strain- and gender-specific benchmarks for distribution of various immune cell subtypes in these organs, we performed immunophenotypic analysis of BM cells and splenocytes from both genders of three commonly used murine strains (C57BL/6NCr, 129/SvHsd, and BALB/cAnNCr). Total neutrophils and splenic macrophages were significantly higher in C57BL/6NCr, whereas total B cells were lower. Within C57BL/6NCr female mice, BM B cells were elevated with respect to the males whereas splenic mDCs and splenic neutrophils were reduced. Within BALB/cAnNCr male mice, BM CD4+ Tregs were elevated with respect to the other strains. Furthermore, in male BALB/cAnNCr mice, NK cells were elevated with respect to the other strains in both BM and spleen. Splenic CD4+ Tregs and splenic CD8+ T cells were reduced in male BALB/c mice in comparison to female mice. Bone marrow CD4+ T cells and mDCs were significantly increased in 129/SvHsd whereas splenic CD8+ T cells were reduced. In general, males exhibited higher immature myeloid cells, macrophages, and NK cells. To our knowledge, this study provides a first attempt to systematically establish organ-specific benchmarks on immune cells in studies involving these mouse strains.
Collapse
|
41
|
Hoffmann HM, Larder R, Lee JS, Hu RJ, Trang C, Devries BM, Clark DD, Mellon PL. Differential CRE Expression in Lhrh-cre and GnRH-cre Alleles and the Impact on Fertility in Otx2-Flox Mice. Neuroendocrinology 2019; 108:328-342. [PMID: 30739114 PMCID: PMC6753941 DOI: 10.1159/000497791] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2018] [Accepted: 02/06/2019] [Indexed: 12/12/2022]
Abstract
There is an increasing trend in studies utilizing cell-specific deletion of genes through conditional gene deletion by CRE recombination. Despite numerous advantages, this strategy also has limitations such as ectopic CRE-expression and germline recombination. Two commonly used gonadotropin-releasing hormone (Gnrh)-driven CRE-expressing mice both target GnRH neurons. However, a direct comparison of the cells targeted and their phenotypic outcome have not yet been presented. To compare where recombination takes place, we crossed the Gnrh-cre and Lhrh-cre lines with the Rosa26-LacZ reporter mouse. Lhrh-cre allowed recombination of the Rosa26-LacZ gene in ∼700 cells, which is comparable to the GnRH neuronal population. Surprisingly, there were > 20 times more LacZ expressing cells in the adult Gnrh-cre:Rosa26-LacZ than the Lhrh-cre:Rosa26-LacZ brain. The greatest differences in targeting of the Gnrh-cre and Lhrh-cre lines were found in the septum, the suprachiasmatic nucleus, and the septohypothalamic area. This difference in cells targeted was present from embryonic day 12. A prior study using the Gnrh-cre to delete the transcription factor Otx2 found fewer GnRH neurons, leading to male and female subfertility. To recapitulate this study, we performed a fertility assay in Otx2:Lhrh-cre mice. We confirmed the requirement for Otx2 in GnRH neuron development, fertility and correct gonadotropin hormone release in Otx2:Lhrh-cre males, but the subfertility was more modest than in Otx2:Gnrh-cre and absent in female Otx2:Lhrh-cre. This suggests that ectopic expression of Gnrh-cre contributes to the reproductive phenotype observed. Finally, the Cre alleles caused germline recombination of the flox allele when transmitted from either parent, generating embryonic lethal knock-out offspring, producing smaller live litters.
Collapse
Affiliation(s)
- Hanne M Hoffmann
- Department of Obstetrics and Gynecology and Reproductive Sciences, Center for Reproductive Science and Medicine, University of California, San Diego, California, USA
- Department of Animal Science, Michigan State University, East Lansing, Michigan, USA
| | - Rachel Larder
- Department of Obstetrics and Gynecology and Reproductive Sciences, Center for Reproductive Science and Medicine, University of California, San Diego, California, USA
| | - Jessica S Lee
- Department of Obstetrics and Gynecology and Reproductive Sciences, Center for Reproductive Science and Medicine, University of California, San Diego, California, USA
| | - Rachael J Hu
- Department of Obstetrics and Gynecology and Reproductive Sciences, Center for Reproductive Science and Medicine, University of California, San Diego, California, USA
| | - Crystal Trang
- Department of Obstetrics and Gynecology and Reproductive Sciences, Center for Reproductive Science and Medicine, University of California, San Diego, California, USA
| | - Brooke M Devries
- Department of Animal Science, Michigan State University, East Lansing, Michigan, USA
| | - Daniel D Clark
- Department of Obstetrics and Gynecology and Reproductive Sciences, Center for Reproductive Science and Medicine, University of California, San Diego, California, USA
| | - Pamela L Mellon
- Department of Obstetrics and Gynecology and Reproductive Sciences, Center for Reproductive Science and Medicine, University of California, San Diego, California, USA,
| |
Collapse
|
42
|
Ghosh A, Sarkar S, Banerjee S, Behbod F, Tawfik O, McGregor D, Graff S, Banerjee SK. MIND model for triple-negative breast cancer in syngeneic mice for quick and sequential progression analysis of lung metastasis. PLoS One 2018; 13:e0198143. [PMID: 29813119 PMCID: PMC5973560 DOI: 10.1371/journal.pone.0198143] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 05/14/2018] [Indexed: 12/25/2022] Open
Abstract
Mouse models of breast cancer with specific molecular subtypes (e.g., ER or HER2 positive) in an immunocompetent or an immunocompromised environment significantly contribute to our understanding of cancer biology, despite some limitations, and they give insight into targeted therapies. However, an ideal triple-negative breast cancer (TNBC) mouse model is lacking. What has been missing in the TNBC mouse model is a sequential progression of the disease in an essential native microenvironment. This notion inspired us to develop a TNBC-model in syngeneic mice using a mammary intraductal (MIND) method. To achieve this goal, Mvt-1and 4T1 TNBC mouse cell lines were injected into the mammary ducts via nipples of FVB/N mice and BALB/c wild-type immunocompetent mice, respectively. We established that the TNBC-MIND model in syngeneic mice could epitomize all breast cancer progression stages and metastasis into the lungs via lymphatic or hematogenous dissemination within four weeks. Collectively, the syngeneic mouse-TNBC-MIND model may serve as a unique platform for further investigation of the underlying mechanisms of TNBC growth and therapies.
Collapse
Affiliation(s)
- Arnab Ghosh
- Cancer Research Unit, VA Medical Center, Kansas City, Missouri, United States of America
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Sandipto Sarkar
- Cancer Research Unit, VA Medical Center, Kansas City, Missouri, United States of America
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Snigdha Banerjee
- Cancer Research Unit, VA Medical Center, Kansas City, Missouri, United States of America
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Fariba Behbod
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Ossama Tawfik
- Saint Luke’s Hospital of Kansas City, Kansas City, Missouri, United States of America
| | - Douglas McGregor
- Cancer Research Unit, VA Medical Center, Kansas City, Missouri, United States of America
- Pathology Department, VA Medical Center, Kansas City, Missouri, United States of America
| | - Stephanie Graff
- Cancer Research Unit, VA Medical Center, Kansas City, Missouri, United States of America
- Sarah Cannon Cancer Center at HCA Midwest Health, Overland Park, Kansas, United States of America
| | - Sushanta K. Banerjee
- Cancer Research Unit, VA Medical Center, Kansas City, Missouri, United States of America
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, Kansas, United States of America
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas, United States of America
- * E-mail: ,
| |
Collapse
|
43
|
Jameson SC, Masopust D. What Is the Predictive Value of Animal Models for Vaccine Efficacy in Humans? Reevaluating the Potential of Mouse Models for the Human Immune System. Cold Spring Harb Perspect Biol 2018; 10:cshperspect.a029132. [PMID: 28348039 DOI: 10.1101/cshperspect.a029132] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Much of what we understand about immunology, including the response to vaccines, come from studies in mice because they provide many practical advantages compared with research in higher mammals and humans. Nevertheless, modalities for preventing or treating disease do not always translate from mouse to humans, which has led to increasing scrutiny of the continued merits of mouse research. Here, we summarize the pros and cons of current laboratory mouse models for immunology research and discuss whether overreliance on nonphysiological, ultra-hygienic animal husbandry approaches has limited the ultimate translation potential of mouse-derived data to humans. Alternative approaches are discussed that may extend the use of the mouse model for preclinical studies.
Collapse
Affiliation(s)
- Stephen C Jameson
- University of Minnesota, Center for Immunology, Minneapolis, Minnesota 55414
| | - David Masopust
- University of Minnesota, Center for Immunology, Minneapolis, Minnesota 55414
| |
Collapse
|
44
|
Abedinpour P, Baron VT, Chrastina A, Rondeau G, Pelayo J, Welsh J, Borgström P. Plumbagin improves the efficacy of androgen deprivation therapy in prostate cancer: A pre-clinical study. Prostate 2017; 77:1550-1562. [PMID: 28971491 DOI: 10.1002/pros.23428] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Accepted: 08/28/2017] [Indexed: 11/08/2022]
Abstract
BACKGROUND Plumbagin is a candidate drug for the treatment of prostate cancer. Previous observations indicated that it may improve the efficacy of androgen deprivation therapy (ADT). This study evaluates the effectiveness of treatment with combinations of plumbagin and alternative strategies for ADT in mouse models of prostate cancer to support its clinical use. METHODS Plumbagin was administered per oral in a new sesame oil formulation. Standard toxicology studies were performed in rats. For tumor growth studies, mouse prostate cancer cell spheroids were placed on top of grafted prostate tissue in a dorsal chamber and allowed to form tumors. Mice were separated in various treatment groups and tumor size was measured over time by intra-vital microscopy. Survival studies were done in mice after injection of prostate cancer cells in the prostate of male animals. Androgen receptor (AR) levels were analyzed by Western blot from prostate cancer cells treated with plumbagin. RESULTS Plumbagin caused a decrease in AR levels in vitro. In mice, plumbagin at 1 mg/kg in sesame oil displayed low toxicity and caused a 50% tumor regression when combined with castration. The combination of plumbagin with various forms of chemical ADT including treatment with a GnRH receptor agonist, a GnRH receptor antagonist, or CYP17A1 inhibitors, outperformed ADT alone, increasing mouse survival compared to the standard regimen of castration alone. In contrast, the combination of plumbagin with AR antagonists, such as bicalutamide and enzalutamide, showed no improvement over AR antagonists alone. Thus, plumbagin is effective in combination with drugs that prevent the synthesis of testosterone or its conversion to dihydrotestosterone, but not with drugs that bind to AR. CONCLUSION Plumbagin significantly improves the effect of ADT drugs currently used in the clinic, with few side effects in mice.
Collapse
Affiliation(s)
- Parisa Abedinpour
- Vaccine Research Institute of San Diego (VRISD), San Diego Science Center, San Diego, California
| | - Véronique T Baron
- Vaccine Research Institute of San Diego (VRISD), San Diego Science Center, San Diego, California
| | - Adrian Chrastina
- Vaccine Research Institute of San Diego (VRISD), San Diego Science Center, San Diego, California
| | - Gaelle Rondeau
- Vaccine Research Institute of San Diego (VRISD), San Diego Science Center, San Diego, California
| | - Jennifer Pelayo
- Vaccine Research Institute of San Diego (VRISD), San Diego Science Center, San Diego, California
| | - John Welsh
- Vaccine Research Institute of San Diego (VRISD), San Diego Science Center, San Diego, California
| | - Per Borgström
- Vaccine Research Institute of San Diego (VRISD), San Diego Science Center, San Diego, California
| |
Collapse
|
45
|
Martin MD, Danahy DB, Hartwig SM, Harty JT, Badovinac VP. Revealing the Complexity in CD8 T Cell Responses to Infection in Inbred C57B/6 versus Outbred Swiss Mice. Front Immunol 2017; 8:1527. [PMID: 29213267 PMCID: PMC5702636 DOI: 10.3389/fimmu.2017.01527] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 10/27/2017] [Indexed: 12/30/2022] Open
Abstract
Recent work has suggested that current mouse models may underrepresent the complexity of human immune responses. While most mouse immunology studies utilize inbred mouse strains, it is unclear if conclusions drawn from inbred mice can be extended to all mouse strains or generalized to humans. We recently described a “surrogate activation marker” approach that could be used to track polyclonal CD8 T cell responses in inbred and outbred mice and noted substantial discord in the magnitude and kinetics of CD8 T cell responses in individual outbred mice following infection. However, how the memory CD8 T cell response develops following infection and the correlates of memory CD8 T cell-mediated protection against re-infection in outbred mice remains unknown. In this study, we investigated development of pathogen-specific memory CD8 T cell responses in inbred C57B/6 and outbred National Institutes of Health Swiss mice following lymphocytic choriomeningitis virus or L. monocytogenes infection. Interestingly, the size of the memory CD8 T cell pool generated and rate of phenotypic progression was considerably more variable in individual outbred compared to inbred mice. Importantly, while prior infection provided both inbred and outbred cohorts of mice with protection against re-infection that was dependent on the dose of primary infection, levels of memory CD8 T cells generated and degree of protection against re-infection did not correlate with primary infection dose in all outbred mice. While variation in CD8 T cell responses to infection is not entirely surprising due to the genetic diversity present, analysis of infection-induced immunity in outbred hosts may reveal hidden complexity in CD8 T cell responses in genetically diverse populations and might help us further bridge the gap between mouse and human studies.
Collapse
Affiliation(s)
- Matthew D Martin
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Derek B Danahy
- Interdisciplinary Graduate Program in Immunology, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Stacey M Hartwig
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA, United States.,Department of Microbiology, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - John T Harty
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA, United States.,Interdisciplinary Graduate Program in Immunology, Carver College of Medicine, University of Iowa, Iowa City, IA, United States.,Department of Microbiology, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Vladimir P Badovinac
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA, United States.,Interdisciplinary Graduate Program in Immunology, Carver College of Medicine, University of Iowa, Iowa City, IA, United States.,Department of Microbiology, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| |
Collapse
|
46
|
Vogt MA, Pfeiffer N, Le Guisquet AM, Brandwein C, Brizard B, Gass P, Belzung C, Chourbaji S. May the use of different background strains 'strain' the stress-related phenotype of GR +/- mice? Behav Brain Res 2017; 335:71-79. [PMID: 28782590 DOI: 10.1016/j.bbr.2017.07.037] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 07/21/2017] [Accepted: 07/25/2017] [Indexed: 01/06/2023]
Abstract
Genetically altered mice are available on different background strains. While respective backcrosses are often performed for pragmatic reasons, e.g. references, comparability, or existing protocols, the interaction between the mutations per se and the background strain often remains a neglected factor. The heterozygous mutation of the glucocorticoid receptor gene (GR) represents a well-examined model for depressive-like behavior in mice. To address the question in how far a robust depressive-like phenotype on a distinct background strain may allow a generalized conclusion, we analyzed respective phenotypes in two commonly used inbred strains: i.) C57BL/6N and ii.) BALB/c. Beside the use of different genetic models, we also extended our approach by applying two alternative paradigms to induce a depressive-like phenotype. Our study therefore comprised the model of 'unpredictable chronic mild stress' (UCMS) for four weeks and 'learned helplessness' (LH), which were used to study the role of GR, a key player in the development of depression. In the course of the experiment two cohorts of male GR+/- mice on either C57BL/6N or BALB/c background strain underwent a behavioral test battery to assess basal and depressive-like features. While both stress paradigms were functional in inducing depressive-like changes, the results were strictly strain-dependent. The genetic consequences became even more obvious under non-stress conditions with significant effects detected in BALB/c mice, which indicates a different basal stress predisposition due to differences in the genetic background.
Collapse
Affiliation(s)
- Miriam A Vogt
- Interfaculty Biomedical Research Facility, University of Heidelberg, Heidelberg, Germany; Central Institute of Mental Health, RG Animal Models in Psychiatry, Medical Faculty of Mannheim/University of Heidelberg, Mannheim, Germany.
| | - Natascha Pfeiffer
- Central Institute of Mental Health, RG Animal Models in Psychiatry, Medical Faculty of Mannheim/University of Heidelberg, Mannheim, Germany
| | - Anne Marie Le Guisquet
- Brain & Imaging (INSERM - UMR 930), Université François Rabelais de Tours, Tours, France
| | - Christiane Brandwein
- Central Institute of Mental Health, RG Animal Models in Psychiatry, Medical Faculty of Mannheim/University of Heidelberg, Mannheim, Germany
| | - Bruno Brizard
- Brain & Imaging (INSERM - UMR 930), Université François Rabelais de Tours, Tours, France
| | - Peter Gass
- Central Institute of Mental Health, RG Animal Models in Psychiatry, Medical Faculty of Mannheim/University of Heidelberg, Mannheim, Germany
| | - Catherine Belzung
- Brain & Imaging (INSERM - UMR 930), Université François Rabelais de Tours, Tours, France
| | - Sabine Chourbaji
- Interfaculty Biomedical Research Facility, University of Heidelberg, Heidelberg, Germany; Central Institute of Mental Health, RG Animal Models in Psychiatry, Medical Faculty of Mannheim/University of Heidelberg, Mannheim, Germany
| |
Collapse
|
47
|
Masopust D, Sivula CP, Jameson SC. Of Mice, Dirty Mice, and Men: Using Mice To Understand Human Immunology. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2017; 199:383-388. [PMID: 28696328 PMCID: PMC5512602 DOI: 10.4049/jimmunol.1700453] [Citation(s) in RCA: 208] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Accepted: 04/28/2017] [Indexed: 12/29/2022]
Abstract
Mouse models have enabled breakthroughs in our understanding of the immune system, but it has become increasingly popular to emphasize their shortcomings when translating observations to humans. This review provides a brief summary of mouse natural history, husbandry, and the pros and cons of pursuing basic research in mice versus humans. Opportunities are discussed for extending the predictive translational value of mouse research, with an emphasis on exploitation of a "dirty" mouse model that better mimics the diverse infectious history that is typical of most humans.
Collapse
Affiliation(s)
- David Masopust
- Department of Microbiology and Immunology, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455;
| | - Christine P Sivula
- Research Animal Resources, University of Minnesota, Minneapolis, MN 55455; and
| | - Stephen C Jameson
- Department of Laboratory Medicine and Pathology, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455
| |
Collapse
|
48
|
Signal peptide peptidase and SPP-like proteases - Possible therapeutic targets? BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2017. [PMID: 28624439 DOI: 10.1016/j.bbamcr.2017.06.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Signal peptide peptidase (SPP) and the four homologous SPP-like proteases SPPL2a, SPPL2b, SPPL2c and SPPL3 are GxGD-type intramembrane-cleaving proteases (I-CLIPs). In addition to divergent subcellular localisations, distinct differences in the mechanistic properties and substrate requirements of individual family members have been unravelled. SPP/SPPL proteases employ a catalytic mechanism related to that of the γ-secretase complex. Nevertheless, differential targeting of SPP/SPPL proteases and γ-secretase by inhibitors has been demonstrated. Furthermore, also within the SPP/SPPL family significant differences in the sensitivity to currently available inhibitory compounds have been reported. Though far from complete, our knowledge on pathophysiological functions of SPP/SPPL proteases, in particular based on studies in mice, has been significantly increased over the last years. Based on this, inhibition of distinct SPP/SPPL proteases has been proposed as a novel therapeutic concept e.g. for the treatment of autoimmunity and viral or protozoal infections, as we will discuss in this review. This article is part of a Special Issue entitled: Proteolysis as a Regulatory Event in Pathophysiology edited by Stefan Rose-John.
Collapse
|
49
|
Saluja R, Kumar A, Jain M, Goel SK, Jain A. Role of Sphingosine-1-Phosphate in Mast Cell Functions and Asthma and Its Regulation by Non-Coding RNA. Front Immunol 2017; 8:587. [PMID: 28588581 PMCID: PMC5439123 DOI: 10.3389/fimmu.2017.00587] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Accepted: 05/03/2017] [Indexed: 01/07/2023] Open
Abstract
Sphingolipid metabolites are emerging as important signaling molecules in allergic diseases specifically asthma. One of the sphingolipid metabolite, sphingosine-1-phosphate (S1P), is involved in cell differentiation, proliferation, survival, migration, and angiogenesis. In the allergic diseases, alteration of S1P levels influences the differentiation and responsiveness of mast cells (MCs). S1P is synthesized by two sphingosine kinases (SphKs), sphingosine kinase 1, and sphingosine kinase 2. Engagement of IgE to the FcεRI receptor induces the activation of both the SphKs and generates S1P. Furthermore, SphKs are also essential to FcεRI-mediated MC activation. Activated MCs export S1P into the extracellular space and causes inflammatory response and tissue remodeling. S1P signaling has dual role in allergic responses. Activation of SphKs and secretion of S1P are required for MC activation; however, S1P signaling plays a vital role in the recovery from anaphylaxis. Several non-coding RNAs have been shown to play a crucial role in controlling the MC-associated inflammatory and allergic responses. Thus, S1P signaling pathway and its regulation by non-coding RNA could be explored as an exciting potential therapeutic target for asthma and other MC-associated diseases.
Collapse
Affiliation(s)
- Rohit Saluja
- Department of Biochemistry, All India Institute of Medical Sciences, Bhopal, India
| | - Ashok Kumar
- Department of Biochemistry, All India Institute of Medical Sciences, Bhopal, India
| | - Manju Jain
- Centre for Biochemistry and Microbial Sciences, Central University of Punjab, Bathinda, India
| | - Sudhir K Goel
- Department of Biochemistry, All India Institute of Medical Sciences, Bhopal, India
| | - Aklank Jain
- Centre for Animal Sciences, Central University of Punjab, Bathinda, India
| |
Collapse
|
50
|
Mitochondrial Calcium Handling in Physiology and Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 982:25-47. [PMID: 28551780 DOI: 10.1007/978-3-319-55330-6_2] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Calcium (Ca2+) accumulation inside mitochondria represents a pleiotropic signal controlling a wide range of cellular functions, including key metabolic pathways and life/death decisions. This phenomenon has been first described in the 1960s, but the identity of the molecules controlling this process remained a mystery until just few years ago, when both mitochondrial Ca2+ uptake and release systems were genetically dissected. This finally opened the possibility to develop genetic models to directly test the contribution of mitochondrial Ca2+ homeostasis to cellular functions. Here we summarize our current understanding of the molecular machinery that controls mitochondrial Ca2+ handling and critically evaluate the physiopathological role of mitochondrial Ca2+ signaling, based on recent evidences obtained through in vitro and in vivo models.
Collapse
|