1
|
Morais JAV, Barros PHA, Brigido MDM, Marina CL, Bocca A, Mariano ADLES, Souza PEND, Paiva KLR, Simões MM, Bao SN, Camargo LC, Longo JPF, Morais AAC, Azevedo RBD, Fonseca MJP, Muehlmann LA. Direct and Abscopal Antitumor Responses Elicited by AlPcNE-Mediated Photodynamic Therapy in a Murine Melanoma Model. Pharmaceutics 2024; 16:1177. [PMID: 39339213 PMCID: PMC11435272 DOI: 10.3390/pharmaceutics16091177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 08/19/2024] [Accepted: 08/25/2024] [Indexed: 09/30/2024] Open
Abstract
Melanoma, the most aggressive form of skin cancer, presents a major clinical challenge due to its tendency to metastasize and recalcitrance to traditional therapies. Despite advances in surgery, chemotherapy, and radiotherapy, the outlook for advanced melanoma remains bleak, reinforcing the urgent need for more effective treatments. Photodynamic therapy (PDT) has emerged as a promising alternative, leading to targeted tumor destruction with minimal harm to surrounding tissues. In this study, the direct and abscopal antitumor effects of PDT in a bilateral murine melanoma model were evaluated. Although only one of the two tumors was treated, effects were observed in both. Our findings revealed significant changes in systemic inflammation and alterations in CD4+ and CD8+ T cell populations in treated groups, as evidenced by blood analyses and flow cytometry. High-throughput RNA sequencing (RNA-Seq) further unveiled shifts in gene expression profiles in both treated and untreated tumors. This research sheds light on the novel antitumor and abscopal effects of nanoemulsion of aluminum chloride phthalocyanine (AlPcNE)-mediated PDT in melanoma, highlighting the potential of different PDT protocols to modulate immune responses and to achieve more effective and targeted cancer treatments.
Collapse
Affiliation(s)
- José Athayde Vasconcelos Morais
- Laboratory of Nanoscience and Immunology, Faculty of Ceilandia, University of Brasilia Ceilandia Sul, Brasilia 72220-275, DF, Brazil
- Laboratory of Gene Regulation and Mutagenesis, Department of Genetics and Morphology, Institute of Biological Sciences, University of Brasilia, Brasilia 70910-900, DF, Brazil
| | - Pedro H A Barros
- Laboratory of Molecular Immunology, Department of Cell Biology, Institute of Biological Sciences, University of Brasilia, Brasilia 70910-900, DF, Brazil
| | - Marcelo de Macedo Brigido
- Laboratory of Molecular Immunology, Department of Cell Biology, Institute of Biological Sciences, University of Brasilia, Brasilia 70910-900, DF, Brazil
| | - Clara Luna Marina
- Laboratory of Applied Immunology, Institute of Biology Sciences, University of Brasilia, Brasilia 70910-900, DF, Brazil
| | - Anamelia Bocca
- Laboratory of Applied Immunology, Institute of Biology Sciences, University of Brasilia, Brasilia 70910-900, DF, Brazil
| | - André de Lima E Silva Mariano
- Laboratory for Softwares and Physics Instrumentation Development, Institute of Physics, University of Brasilia, Brasilia 70910-900, DF, Brazil
| | - Paulo E N de Souza
- Laboratory for Softwares and Physics Instrumentation Development, Institute of Physics, University of Brasilia, Brasilia 70910-900, DF, Brazil
| | - Karen L R Paiva
- Laboratory of Microscopy and Microanalysis, Department of Cell Biology, Institute of Biological Sciences, University of Brasilia, Brasilia 70910-900, DF, Brazil
| | - Marina Mesquita Simões
- Laboratory of Microscopy and Microanalysis, Department of Cell Biology, Institute of Biological Sciences, University of Brasilia, Brasilia 70910-900, DF, Brazil
| | - Sonia Nair Bao
- Laboratory of Microscopy and Microanalysis, Department of Cell Biology, Institute of Biological Sciences, University of Brasilia, Brasilia 70910-900, DF, Brazil
| | - Luana C Camargo
- Laboratory of Nanoscience and Nanobiotechnology, Department of Genetics and Morphology, Institute of Biological Sciences, University of Brasilia, Brasilia 70910-900, DF, Brazil
| | - João P Figueiró Longo
- Laboratory of Nanoscience and Nanobiotechnology, Department of Genetics and Morphology, Institute of Biological Sciences, University of Brasilia, Brasilia 70910-900, DF, Brazil
| | - Amanda Alencar Cabral Morais
- Laboratory of Nanoscience and Nanobiotechnology, Department of Genetics and Morphology, Institute of Biological Sciences, University of Brasilia, Brasilia 70910-900, DF, Brazil
| | - Ricardo B de Azevedo
- Laboratory of Nanoscience and Nanobiotechnology, Department of Genetics and Morphology, Institute of Biological Sciences, University of Brasilia, Brasilia 70910-900, DF, Brazil
| | - Marcio J P Fonseca
- Laboratory of Gene Regulation and Mutagenesis, Department of Genetics and Morphology, Institute of Biological Sciences, University of Brasilia, Brasilia 70910-900, DF, Brazil
| | - Luis A Muehlmann
- Laboratory of Nanoscience and Immunology, Faculty of Ceilandia, University of Brasilia Ceilandia Sul, Brasilia 72220-275, DF, Brazil
| |
Collapse
|
2
|
Prentiss RL, Bollinger BL, Lamont KA, Gaston KN, Fletcher CA, Williams MD, Atkins HM, Galex IA. Comparison of Novel and Traditional Bleeding Techniques in Neonatal and Juvenile Mice. JOURNAL OF THE AMERICAN ASSOCIATION FOR LABORATORY ANIMAL SCIENCE : JAALAS 2024; 63:333-342. [PMID: 38514171 PMCID: PMC11193428 DOI: 10.30802/aalas-jaalas-23-000116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 01/24/2024] [Accepted: 02/13/2024] [Indexed: 03/23/2024]
Abstract
Blood collection is frequently used for neonatal and juvenile mice in toxicology, developmental, and immunology studies and is often a terminal procedure. However, the use of nonterminal blood collection techniques, including the submandibular and the submental collection techniques described for adult mice, may offer opportunities to reduce animal numbers and refine current methods. The use of the submental technique has not been described for neonatal or juvenile mice. In this study, we compared the submental and submandibular blood collection techniques to determine their suitability for use in neonatal and juvenile mice. Male and female CD1 mice, ages 7, 14, 21, and 28 d, were randomized by sex into submental (n = 16), submandibular (n = 16), or control (n = 8) groups. Each mouse was weighed, bled per its assigned group (or only restrained in the case of control mice), and then decapitated without anesthesia for terminal blood collection. Blood collection volume and corticosterone concentrations were measured. The 2 methods showed significant differences in the volume of blood collected at ages 14 and 28, with the submandibular technique yielding significantly higher volumes. No significant differences were detected in corticosterone levels between the 2 techniques based on age or sex. A subset of mice (n = 8, 2 per age group) were bled via submental or submandibular technique and were evaluated 48 h later for gross and histopathologic evidence of trauma. Seven of the 8 mice showed expected inflammation and healing at the collection sites, with 4 mice having embedded strands of fur in the tissue. These data indicate that the submental blood collection is a viable method for nonterminal blood collection method in neonatal and juvenile mice, especially when smaller amounts of blood are needed.
Collapse
Affiliation(s)
- Rebecca L Prentiss
- Division of Comparative Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Brooke L Bollinger
- Division of Comparative Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Katherine A Lamont
- Division of Comparative Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Kimberly N Gaston
- Division of Comparative Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Craig A Fletcher
- Division of Comparative Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Morika D Williams
- Division of Comparative Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- UNC Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; and
- Thurston Arthritis Research Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Hannah M Atkins
- Division of Comparative Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Ilana A Galex
- Division of Comparative Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| |
Collapse
|
3
|
Tracy GC, Huang KY, Hong YT, Ding S, Noblet HA, Lim KH, Kim EC, Chung HJ, Kong H. Intracerebral Nanoparticle Transport Facilitated by Alzheimer Pathology and Age. NANO LETTERS 2023; 23:10971-10982. [PMID: 37991895 PMCID: PMC11404402 DOI: 10.1021/acs.nanolett.3c03222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2023]
Abstract
Nanoparticles have emerged as potential transporters of drugs targeting Alzheimer's disease (AD), but their design should consider the blood-brain barrier (BBB) integrity and neuroinflammation of the AD brain. This study presents that aging is a significant factor for the brain localization and retention of nanoparticles, which we engineered to bind with reactive astrocytes and activated microglia. We assembled 200 nm-diameter particles using a block copolymer of poly(lactic-co-glycolic acid) (PLGA) and CD44-binding hyaluronic acid (HA). The resulting PLGA-b-HA nanoparticles displayed increased binding to CD44-expressing reactive astrocytes and activated microglia. Upon intravascular injection, nanoparticles were localized to the hippocampi of both APP/PS1 AD model mice and their control littermates at 13-16 months of age due to enhanced transvascular transport through the leaky BBB. No particles were found in the hippocampi of young adult mice. These findings demonstrate the brain localization of nanoparticles due to aging-induced BBB breakdown regardless of AD pathology.
Collapse
Affiliation(s)
- Gregory C Tracy
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Kai-Yu Huang
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Yu-Tong Hong
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Shengzhe Ding
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Hayden A Noblet
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Ki H Lim
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Eung Chang Kim
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Hee Jung Chung
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Hyunjoon Kong
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seongbuk-gu, Seoul 02841, South Korea
| |
Collapse
|
4
|
Individualized Housing Modifies the Immune–Endocrine System in CD1 Adult Male Mice. Animals (Basel) 2023; 13:ani13061026. [PMID: 36978567 PMCID: PMC10044133 DOI: 10.3390/ani13061026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 03/08/2023] [Accepted: 03/09/2023] [Indexed: 03/14/2023] Open
Abstract
In the last years, different research groups have made considerable efforts to improve the care and use of animals in research. Mice (Mus musculus) are the most widely used animal species in research in the European Union and are sociable and hierarchical creatures. During experiments, researchers tend to individualize males, but no consideration is given to whether this social isolation causes them stress. The aim of this study was, therefore, to explore whether 4 weeks of social isolation could induce changes in different physiological parameters in adult Crl:CD1(ICR) (CD1) males, which may interfere with experimental results. Body weight, blood cells, and fecal corticosterone metabolites levels were the analyzed parameters. Blood and fecal samples were collected at weeks 1 and 4 of the experimental procedure. Four weeks of single housing produced a significant time-dependent decrease in monocytes and granulocytes. Fecal corticosterone metabolite levels were higher in single-housed mice after 1 week and then normalized after 4 weeks of isolation. Body weight, red blood cells, and platelets remained unchanged in both groups during this period. We can, therefore, conclude that social isolation affects some immune and endocrine parameters, and that this should be taken into account in the interpretation of research data.
Collapse
|
5
|
Chung Y, Desiraju S, Namachivayam K, Guzman P, He L, MohanKumar K. Hematological changes in neonatal mice with phlebotomy-induced anemia. Pediatr Res 2022; 92:1575-1579. [PMID: 35322186 PMCID: PMC9500113 DOI: 10.1038/s41390-022-02023-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 11/18/2021] [Accepted: 03/06/2022] [Indexed: 12/30/2022]
Abstract
BACKGROUND Anemia is a nearly universal diagnosis in preterm infants, caused by phlebotomy, and exacerbated by the underlying erythropoietic immaturity. Newborn infants are exposed to the unique stressor of fetal-to-neonatal transition, which requires significant adaptation ex utero. Accordingly, the preterm infant's response to anemia may alter the ability to confront underlying illness. This study utilized our preclinical mouse model of phlebotomy-induced anemia (PIA) to comprehensively investigate associated hematological changes. METHODS C57BL/6 mice were subjected to timed phlebotomy between postnatal days 2--10 to induce severe anemia. Complete blood counts were determined by the Sysmex XT-2000iV analyzer. RESULTS Anemic pups showed a gradual reduction of RBC and hemoglobin (Hb) and increased reticulocyte (RET) counts and red cell distribution width (RDW), however, with reduced RET-Hb from postnatal day (P) of 4 onwards. Elevated levels of high fluorescent RET and immature reticulocyte fraction (IRF) were noted in anemic mouse pups, but low and medium fluorescent RET were reduced. Also, the reduction of mean corpuscular volume (MCV), mean corpuscular hemoglobin (MCH), and mean corpuscular hemoglobin concentration (MCHC) were noted in anemic pups. No changes were seen in lymphocytes, but monocytes and neutrophils were significantly elevated from P4-P6. CONCLUSIONS PIA in mouse pups is associated with hematological changes that may be exacerbating factors in neonatal diseases. IMPACT Anemia is common and often severe in premature infants. Investigation of hematological parameters in settings of preclinical anemia may be an index of therapeutic strategies. Preclinical model evaluating the effects of neonatal anemia on the remainder of complete blood count. Detailed time kinetic phlebotomy-induced anemic mice enable us to study the impact on developmental delays in erythropoiesis and possible strategic intervention. Hematological effects of severe anemia in mice might provide insight on how best to investigate anemia in preterm infants.
Collapse
Affiliation(s)
- Yerin Chung
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Suneetha Desiraju
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | | | - Pierre Guzman
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Ling He
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- Department of Pharmacology & Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Krishnan MohanKumar
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA.
| |
Collapse
|
6
|
Peripheral immune cells and perinatal brain injury: a double-edged sword? Pediatr Res 2022; 91:392-403. [PMID: 34750522 PMCID: PMC8816729 DOI: 10.1038/s41390-021-01818-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 08/24/2021] [Accepted: 09/14/2021] [Indexed: 01/07/2023]
Abstract
Perinatal brain injury is the leading cause of neurological mortality and morbidity in childhood ranging from motor and cognitive impairment to behavioural and neuropsychiatric disorders. Various noxious stimuli, including perinatal inflammation, chronic and acute hypoxia, hyperoxia, stress and drug exposure contribute to the pathogenesis. Among a variety of pathological phenomena, the unique developing immune system plays an important role in the understanding of mechanisms of injury to the immature brain. Neuroinflammation following a perinatal insult largely contributes to evolution of damage to resident brain cells, but may also be beneficial for repair activities. The present review will focus on the role of peripheral immune cells and discuss processes involved in neuroinflammation under two frequent perinatal conditions, systemic infection/inflammation associated with encephalopathy of prematurity (EoP) and hypoxia/ischaemia in the context of neonatal encephalopathy (NE) and stroke at term. Different immune cell subsets in perinatal brain injury including their infiltration routes will be reviewed and critical aspects such as sex differences and maturational stage will be discussed. Interactions with existing regenerative therapies such as stem cells and also potentials to develop novel immunomodulatory targets are considered. IMPACT: Comprehensive summary of current knowledge on the role of different immune cell subsets in perinatal brain injury including discussion of critical aspects to be considered for development of immunomodulatory therapies.
Collapse
|
7
|
Adayev T, LaFauci G, Xu W, Dobkin C, Kascsak R, Brown WT, Goodman JH. Development of a Quantitative FMRP Assay for Mouse Tissue Applications. Genes (Basel) 2021; 12:genes12101516. [PMID: 34680911 PMCID: PMC8535242 DOI: 10.3390/genes12101516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 09/24/2021] [Accepted: 09/24/2021] [Indexed: 11/20/2022] Open
Abstract
Fragile X syndrome results from the absence of the FMR1 gene product—Fragile X Mental Retardation Protein (FMRP). Fragile X animal research has lacked a reliable method to quantify FMRP. We report the development of an array of FMRP-specific monoclonal antibodies and their application for quantitative assessment of FMRP (qFMRPm) in mouse tissue. To characterize the assay, we determined the normal variability of FMRP expression in four brain structures of six different mouse strains at seven weeks of age. There was a hierarchy of FMRP expression: neocortex > hippocampus > cerebellum > brainstem. The expression of FMRP was highest and least variable in the neocortex, whereas it was most variable in the hippocampus. Male C57Bl/6J and FVB mice were selected to determine FMRP developmental differences in the brain at 3, 7, 10, and 14 weeks of age. We examined the four structures and found a developmental decline in FMRP expression with age, except for the brainstem where it remained stable. qFMRPm assay of blood had highest values in 3 week old animals and dropped by 2.5-fold with age. Sex differences were not significant. The results establish qFMRPm as a valuable tool due to its ease of methodology, cost effectiveness, and accuracy.
Collapse
Affiliation(s)
- Tatyana Adayev
- New York State Institute for Basic Research in Developmental Disabilities, New York, NY 10314, USA; (G.L.); (W.X.); (C.D.); (R.K.); (W.T.B.); (J.H.G.)
- Correspondence: ; Tel.: +1-718-494-5314
| | - Giuseppe LaFauci
- New York State Institute for Basic Research in Developmental Disabilities, New York, NY 10314, USA; (G.L.); (W.X.); (C.D.); (R.K.); (W.T.B.); (J.H.G.)
| | - Weimin Xu
- New York State Institute for Basic Research in Developmental Disabilities, New York, NY 10314, USA; (G.L.); (W.X.); (C.D.); (R.K.); (W.T.B.); (J.H.G.)
| | - Carl Dobkin
- New York State Institute for Basic Research in Developmental Disabilities, New York, NY 10314, USA; (G.L.); (W.X.); (C.D.); (R.K.); (W.T.B.); (J.H.G.)
| | - Richard Kascsak
- New York State Institute for Basic Research in Developmental Disabilities, New York, NY 10314, USA; (G.L.); (W.X.); (C.D.); (R.K.); (W.T.B.); (J.H.G.)
| | - W. Ted Brown
- New York State Institute for Basic Research in Developmental Disabilities, New York, NY 10314, USA; (G.L.); (W.X.); (C.D.); (R.K.); (W.T.B.); (J.H.G.)
- Perkins Center, University of Sydney Camperdown, Sydney, NSW 2006, Australia
| | - Jeffrey H. Goodman
- New York State Institute for Basic Research in Developmental Disabilities, New York, NY 10314, USA; (G.L.); (W.X.); (C.D.); (R.K.); (W.T.B.); (J.H.G.)
| |
Collapse
|
8
|
Davenport P, Lorenz V, Liu ZJ, Feldman HA, Canas J, Nolton E, Badur CA, Do TMT, Sola-Visner M. Development of gates to measure the immature platelet fraction in C57BL/6J mice using the Sysmex XN-V series multispecies hematology analyzer. J Vet Diagn Invest 2021; 33:913-919. [PMID: 34218748 DOI: 10.1177/10406387211027899] [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: 11/17/2022] Open
Abstract
The immature platelet fraction (IPF) is a measure of newly released platelets, which has been used as a marker of platelet production in multiple human studies but is not widely available in multispecies analyzers. We developed gates to measure the IPF in diluted and undiluted murine blood samples on the Sysmex XN-1000V multispecies hematology analyzer. IPF gates were created using undiluted and diluted (1/10) blood samples obtained from adult and newborn (postnatal day 10, P10) C57BL/6J wild-type (WT) mice, and from 3 murine models of thrombocytopenia: c-MPL-/- mice, which lack the thrombopoietin receptor (hyporegenerative); antibody-mediated thrombocytopenia; and acute inflammation-induced thrombocytopenia. P10 mice were chosen because, at their size, we could consistently obtain (by terminal phlebotomy) the blood volume needed to run an undiluted sample. The undiluted blood IPF gate successfully differentiated between mechanisms of thrombocytopenia in both adult and P10 mice. For diluted samples, 2 IPF gates were generated: a thrombocytopenic (T) gate, which performed well in samples with platelet counts (PCs) <800 × 109/L in adult mice and <500 × 109/L in newborn mice, and a non-thrombocytopenic (NT) gate, which performed well in samples with PCs above these thresholds. PCs and IPFs measured in diluted blood using these gates agreed well with those measured in undiluted blood and had good reproducibility. These diluted gates allow for the accurate measurement of PCs and IPFs in small (10 µL) blood volumes, which can be obtained easily from adult and newborn mice as small as P1 to assess platelet production serially.
Collapse
Affiliation(s)
| | | | | | - Henry A Feldman
- Institutional Centers for Clinical and Translational Research, Boston Children's Hospital, Boston, MA, USA
| | | | | | | | | | | |
Collapse
|
9
|
Feeding Formula Eliminates the Necessity of Bacterial Dysbiosis and Induces Inflammation and Injury in the Paneth Cell Disruption Murine NEC Model in an Osmolality-Dependent Manner. Nutrients 2020; 12:nu12040900. [PMID: 32224880 PMCID: PMC7230818 DOI: 10.3390/nu12040900] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 03/19/2020] [Accepted: 03/24/2020] [Indexed: 01/08/2023] Open
Abstract
Necrotizing enterocolitis (NEC) remains a significant cause of morbidity and mortality in preterm infants. Formula feeding is a risk factor for NEC and osmolality, which is increased by the fortification that is required for adequate growth of the infant, has been suggested as a potential cause. Our laboratory has shown that Paneth cell disruption followed by induction of dysbiosis can induce NEC-like pathology in the absence of feeds. We hypothesized adding formula feeds to the model would exacerbate intestinal injury and inflammation in an osmolality-dependent manner. NEC-like injury was induced in 14-16 day-old C57Bl/6J mice by Paneth cell disruption with dithizone or diphtheria toxin, followed by feeding rodent milk substitute with varying osmolality (250-1491 mOsm/kg H2O). Animal weight, serum cytokines and osmolality, small intestinal injury, and cecal microbial composition were quantified. Paneth cell-disrupted mice fed formula had significant NEC scores compared to controls and no longer required induction of bacterial dysbiosis. Significant increases in serum inflammatory markers, small intestinal damage, and overall mortality were osmolality-dependent and not related to microbial changes. Overall, formula feeding in combination with Paneth cell disruption induced NEC-like injury in an osmolality-dependent manner, emphasizing the importance of vigilance in designing preterm infant feeds.
Collapse
|
10
|
Bednarz A, Lipiński P, Starzyński RR, Tomczyk M, Nowak W, Mucha O, Ogórek M, Pierzchała O, Jończy A, Staroń R, Śmierzchalska J, Rajfur Z, Baster Z, Józkowicz A, Lenartowicz M. Role of the kidneys in the redistribution of heme-derived iron during neonatal hemolysis in mice. Sci Rep 2019; 9:11102. [PMID: 31366967 PMCID: PMC6668426 DOI: 10.1038/s41598-019-47414-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 06/19/2019] [Indexed: 02/06/2023] Open
Abstract
Moderate intravascular hemolysis is a common condition in newborns. It is followed by the accumulation of bilirubin, which is a secondary product of the activity of heme oxygenase-1, an enzyme that catalyzes the breakdown of heme released from disrupted erythrocytes and taken up by hepatic macrophages. Although these cells are a major site of enzymatic heme breakdown in adults, we show here that epithelial cells of proximal tubules in the kidneys perform the functions of both heme uptake and catabolism in mouse neonates. A time-course study examining mouse pups during the neonatal period showed a gradual recovery from hemolysis, and concomitant decreases in the expression of heme-related genes and non-heme iron transporters in the proximal tubules. By adjusting the expression of iron-handling proteins in response to the disappearance of hemolysis in mouse neonates, the kidneys may play a role in the detoxification of iron and contribute to its recirculation from the primary urine to the blood.
Collapse
Affiliation(s)
- Aleksandra Bednarz
- Department of Genetics and Evolution, Institute of Zoology and Biomedical Research, Jagiellonian University, Gronostajowa 9, 30-387, Kraków, Poland
| | - Paweł Lipiński
- Department of Molecular Biology, Institute of Genetics and Animal Breeding, Polish Academy of Sciences, 05-552, Magdalenka, Jastrzębiec, Poland
| | - Rafał R Starzyński
- Department of Molecular Biology, Institute of Genetics and Animal Breeding, Polish Academy of Sciences, 05-552, Magdalenka, Jastrzębiec, Poland
| | - Mateusz Tomczyk
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland
| | - Witold Nowak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland
| | - Olga Mucha
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland
| | - Mateusz Ogórek
- Department of Genetics and Evolution, Institute of Zoology and Biomedical Research, Jagiellonian University, Gronostajowa 9, 30-387, Kraków, Poland
| | - Olga Pierzchała
- Department of Genetics and Evolution, Institute of Zoology and Biomedical Research, Jagiellonian University, Gronostajowa 9, 30-387, Kraków, Poland
| | - Aneta Jończy
- Department of Molecular Biology, Institute of Genetics and Animal Breeding, Polish Academy of Sciences, 05-552, Magdalenka, Jastrzębiec, Poland
| | - Robert Staroń
- Department of Molecular Biology, Institute of Genetics and Animal Breeding, Polish Academy of Sciences, 05-552, Magdalenka, Jastrzębiec, Poland
| | - Julia Śmierzchalska
- Department of Genetics and Evolution, Institute of Zoology and Biomedical Research, Jagiellonian University, Gronostajowa 9, 30-387, Kraków, Poland
| | - Zenon Rajfur
- Department of Molecular and Interfacial Biophysics, Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, Łojasiewicza 11, 30-348, Kraków, Poland
| | - Zbigniew Baster
- Department of Molecular and Interfacial Biophysics, Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, Łojasiewicza 11, 30-348, Kraków, Poland
| | - Alicja Józkowicz
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland
| | - Małgorzata Lenartowicz
- Department of Genetics and Evolution, Institute of Zoology and Biomedical Research, Jagiellonian University, Gronostajowa 9, 30-387, Kraków, Poland.
| |
Collapse
|
11
|
Aurbach K, Spindler M, Haining EJ, Bender M, Pleines I. Blood collection, platelet isolation and measurement of platelet count and size in mice-a practical guide. Platelets 2018; 30:698-707. [PMID: 30346859 DOI: 10.1080/09537104.2018.1528345] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Inherited or acquired disorders of platelet production and function can result in thrombocytopenia and bleeding. Mouse models have proven useful for investigating the mechanisms that underlie these defects in humans. Precise methods for blood withdrawal, platelet isolation and measurement of platelet parameters are key for the generation of reproducible and conclusive data. Here, we provide three different protocols for mouse platelet isolation to encourage research knowledge transfer between experienced laboratories, while at the same time enabling less experienced researchers to implement a protocol that best suits their local expertise and equipment. We also address the issue that reported mouse platelet count and size vary considerably in the literature by investigating different factors that influence these important platelet parameters, namely: 1) genetic background and gender, 2) choice of analysis method (hematological analyzer or flow cytometry), 3) dilution of the blood sample and 4) choice of anticoagulant. The herein presented results and considerations may serve as a practical guide for both experienced and new researchers in the platelet field.
Collapse
Affiliation(s)
- Katja Aurbach
- a Institute of Experimental Biomedicine I, University Hospital and Rudolf Virchow Center , University of Würzburg , Würzburg , Germany
| | - Markus Spindler
- a Institute of Experimental Biomedicine I, University Hospital and Rudolf Virchow Center , University of Würzburg , Würzburg , Germany
| | - Elizabeth J Haining
- b Institute of Cardiovascular Science, College of Medical and Dental Sciences , University of Birmingham, Edgbaston , Birmingham , UK
| | - Markus Bender
- a Institute of Experimental Biomedicine I, University Hospital and Rudolf Virchow Center , University of Würzburg , Würzburg , Germany
| | - Irina Pleines
- a Institute of Experimental Biomedicine I, University Hospital and Rudolf Virchow Center , University of Würzburg , Würzburg , Germany
| |
Collapse
|
12
|
Herz J, Köster C, Crasmöller M, Abberger H, Hansen W, Felderhoff-Müser U, Bendix I. Peripheral T Cell Depletion by FTY720 Exacerbates Hypoxic-Ischemic Brain Injury in Neonatal Mice. Front Immunol 2018; 9:1696. [PMID: 30127782 PMCID: PMC6087766 DOI: 10.3389/fimmu.2018.01696] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 07/10/2018] [Indexed: 11/13/2022] Open
Abstract
Hypoxic-ischemic injury to the developing brain remains a major cause of significant long-term morbidity and mortality. Emerging evidence from neonatal brain injury models suggests a detrimental role for peripheral lymphocytes. The immunomodulatory substance FTY720, a sphingosine-1-phosphate receptor agonist, was shown to reduce adult ischemia-induced neurodegeneration through its lymphopenic mode of action. In the present study, we hypothesized that FTY720 promotes neuroprotection by reducing peripheral lymphocytes and their infiltration into the injured neonatal brain. Term-born equivalent postnatal day 9 C57BL/6 mice were exposed to hypoxia ischemia (HI) followed by a single injection of 1 mg/kg FTY720 or vehicle (0.9% sodium chloride). Brain injury, microglia, and endothelial activation were assessed 7 days post HI using histology and western blot. Peripheral and cerebral leukocyte subsets were analyzed by multichannel flow cytometry. Whether FTY720s’ effects could be attributed to its lymphopenic mode of action was determined in T cell-depleted mice. In contrast to our hypothesis, FTY720 exacerbated HI-induced neuropathology including loss of gray and white matter structures. While microglia and endothelial activation remained unchanged, FTY720 induced a strong and sustained depletion of peripheral T cells resulting in significantly reduced cerebral infiltration of CD4 T cells. CD4 T cell subset analysis revealed that circulating regulatory and effector T cells counts were similarly decreased after FTY720 treatment. However, since neonatal HI per se induces a selective infiltration of Foxp3 positive regulatory T cells compared to Foxp3 negative effector T cells effects of FTY720 on cerebral regulatory T cell infiltration were more pronounced than on effector T cells. Reductions in T lymphocytes, and particularly regulatory T cells coincided with an increased infiltration of innate immune cells, mainly neutrophils and inflammatory macrophages. Importantly anti-CD3-mediated T cell depletion resulted in a similar exacerbation of brain injury, which was not further enhanced by an additional FTY720 treatment. In summary, peripheral T cell depletion by FTY720 resulted in increased infiltration of innate immune cells concomitant to reduced T cell infiltration and exacerbation HI-induced brain injury. This study indicates that neonatal T cells may promote endogenous neuroprotection in the term-born equivalent hypoxic-ischemic brain potentially providing new opportunities for therapeutic intervention.
Collapse
Affiliation(s)
- Josephine Herz
- Department of Pediatrics 1, Neonatology and Experimental Perinatal Neuroscience, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Christian Köster
- Department of Pediatrics 1, Neonatology and Experimental Perinatal Neuroscience, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Marius Crasmöller
- Department of Pediatrics 1, Neonatology and Experimental Perinatal Neuroscience, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Hanna Abberger
- Institute of Medical Microbiology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Wiebke Hansen
- Institute of Medical Microbiology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Ursula Felderhoff-Müser
- Department of Pediatrics 1, Neonatology and Experimental Perinatal Neuroscience, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Ivo Bendix
- Department of Pediatrics 1, Neonatology and Experimental Perinatal Neuroscience, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
13
|
He Q, Su G, Liu K, Zhang F, Jiang Y, Gao J, Liu L, Jiang Z, Jin M, Xie H. Sex-specific reference intervals of hematologic and biochemical analytes in Sprague-Dawley rats using the nonparametric rank percentile method. PLoS One 2017; 12:e0189837. [PMID: 29261747 PMCID: PMC5738108 DOI: 10.1371/journal.pone.0189837] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Accepted: 12/01/2017] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Hematologic and biochemical analytes of Sprague-Dawley rats are commonly used to determine effects that were induced by treatment and to evaluate organ dysfunction in toxicological safety assessments, but reference intervals have not been well established for these analytes. Reference intervals as presently defined for these analytes in Sprague-Dawley rats have not used internationally recommended statistical method nor stratified by sex. Thus, we aimed to establish sex-specific reference intervals for hematologic and biochemical parameters in Sprague-Dawley rats according to Clinical and Laboratory Standards Institute C28-A3 and American Society for Veterinary Clinical Pathology guideline. METHODS Hematology and biochemistry blood samples were collected from 500 healthy Sprague-Dawley rats (250 males and 250 females) in the control groups. We measured 24 hematologic analytes with the Sysmex XT-2100i analyzer, 9 biochemical analytes with the Olympus AU400 analyzer. We then determined statistically relevant sex partitions and calculated reference intervals, including corresponding 90% confidence intervals, using nonparametric rank percentile method. RESULTS We observed that most hematologic and biochemical analytes of Sprague-Dawley rats were significantly influenced by sex. Males had higher hemoglobin, hematocrit, red blood cell count, red cell distribution width, mean corpuscular volume, mean corpuscular hemoglobin, white blood cell count, neutrophils, lymphocytes, monocytes, percentage of neutrophils, percentage of monocytes, alanine aminotransferase, aspartate aminotransferase, and triglycerides compared to females. Females had higher mean corpuscular hemoglobin concentration, plateletcrit, platelet count, eosinophils, percentage of lymphocytes, percentage of eosinophils, creatinine, glucose, total cholesterol and urea compared to males. Sex partition was required for most hematologic and biochemical analytes in Sprague-Dawley rats. We established sex-specific reference intervals, including corresponding 90% confidence intervals, for Sprague-Dawley rats. CONCLUSIONS Understanding the significant discrepancies in hematologic and biochemical analytes between male and female Sprague-Dawley rats provides important insight into physiological effects in test rats. Establishment of locally sex-specific reference intervals allows a more precise evaluation of animal quality and experimental results of Sprague-Dawley rats in our toxicology safety assessment.
Collapse
Affiliation(s)
- Qili He
- Institute of Toxicological Detection, Sichuan Center for Disease Control and Prevention, Chengdu, Sichuan, China
| | - Guoming Su
- Department of Pharmacy and Laboratory, Sichuan Nursing Vocational College, Chengdu, Sichuan, China
| | - Keliang Liu
- Institute of Toxicological Detection, Sichuan Center for Disease Control and Prevention, Chengdu, Sichuan, China
- * E-mail:
| | - Fangcheng Zhang
- Department of Ultrastructural Pathology Center, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Yong Jiang
- Institute of Toxicological Detection, Sichuan Center for Disease Control and Prevention, Chengdu, Sichuan, China
| | - Jun Gao
- Institute of Toxicological Detection, Sichuan Center for Disease Control and Prevention, Chengdu, Sichuan, China
| | - Lida Liu
- Institute of Toxicological Detection, Sichuan Center for Disease Control and Prevention, Chengdu, Sichuan, China
| | - Zhongren Jiang
- Institute of Toxicological Detection, Sichuan Center for Disease Control and Prevention, Chengdu, Sichuan, China
| | - Minwu Jin
- Institute of Toxicological Detection, Sichuan Center for Disease Control and Prevention, Chengdu, Sichuan, China
| | - Huiping Xie
- Institute of Toxicological Detection, Sichuan Center for Disease Control and Prevention, Chengdu, Sichuan, China
| |
Collapse
|
14
|
Lai JCY, Rocha-Ferreira E, Ek CJ, Wang X, Hagberg H, Mallard C. Immune responses in perinatal brain injury. Brain Behav Immun 2017; 63:210-223. [PMID: 27865947 DOI: 10.1016/j.bbi.2016.10.022] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Revised: 10/28/2016] [Accepted: 10/30/2016] [Indexed: 12/13/2022] Open
Abstract
The perinatal period has often been described as immune deficient. However, it has become clear that immune responses in the neonate following exposure to microbes or as a result of tissue injury may be substantial and play a role in perinatal brain injury. In this article we will review the immune cell composition under normal physiological conditions in the perinatal period, both in the human and rodent. We will summarize evidence of the inflammatory responses to stimuli and discuss how neonatal immune activation, both in the central nervous system and in the periphery, may contribute to perinatal hypoxic-ischemic brain injury.
Collapse
Affiliation(s)
- Jacqueline C Y Lai
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Box 432, 405 30 Gothenburg, Sweden
| | - Eridan Rocha-Ferreira
- Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Box 432, 405 30 Gothenburg, Sweden
| | - C Joakim Ek
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Box 432, 405 30 Gothenburg, Sweden
| | - Xiaoyang Wang
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Box 432, 405 30 Gothenburg, Sweden
| | - Henrik Hagberg
- Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Box 432, 405 30 Gothenburg, Sweden
| | - Carina Mallard
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Box 432, 405 30 Gothenburg, Sweden.
| |
Collapse
|
15
|
Namachivayam K, MohanKumar K, Garg L, Torres BA, Maheshwari A. Neonatal mice with necrotizing enterocolitis-like injury develop thrombocytopenia despite increased megakaryopoiesis. Pediatr Res 2017; 81:817-824. [PMID: 28085792 PMCID: PMC5425334 DOI: 10.1038/pr.2017.7] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 11/19/2016] [Indexed: 01/21/2023]
Abstract
BACKGROUND Thrombocytopenia is frequently encountered in infants with necrotizing enterocolitis (NEC). To develop a preclinical model of NEC-related thrombocytopenia, we measured serial platelet counts in 10-d-old (P10) mouse pups with trinitrobenzene sulfonic acid (TNBS)-induced NEC-like injury. We also measured platelet volume indices, immature platelet fraction (IPF), and megakaryocyte number/ploidy in these animals. METHODS Platelet counts, platelet volume indices, and IPF were measured in control (N = 65) and TNBS-treated pups (N = 104) using an automated hematology analyzer. Bone marrow megakaryocyte number, ploidy and CD41 expression were measured by flow cytometry. These findings were confirmed in a small cohort of P3 mice with NEC-like injury. RESULTS Murine pups with TNBS-mediated NEC-like injury developed thrombocytopenia at 15-24 h after exposure to TNBS. Intestinal injury was associated with increased platelet volume indices (mean platelet volume, platelet-to-large cell ratio, and platelet distribution width), and IPF, indicating increased thrombopoiesis. These mice also showed increased megakaryocyte number, ploidy, and CD41 expression, indicating increased megakaryocyte differentiation. CONCLUSION Similar to human NEC, murine NEC-like injury was also associated with decreased platelet counts. There was evidence of increased megakaryocyte differentiation and thrombopoiesis, which favors peripheral consumption of platelets as the likely mechanism of thrombocytopenia in these animals, over decreased platelet production.
Collapse
Affiliation(s)
| | - Krishnan MohanKumar
- Department of Pediatrics, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| | - Lalit Garg
- Faculty of Information and Communication Technology, University of Malta, Msida, Malta
| | - Benjamin A. Torres
- Department of Pediatrics, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| | - Akhil Maheshwari
- Department of Pediatrics, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
- Department of Community and Family Health, College of Public Health, University of South Florida, Tampa, USA
| |
Collapse
|
16
|
White JR, Gong H, Pope B, Schlievert P, McElroy SJ. Paneth-cell-disruption-induced necrotizing enterocolitis in mice requires live bacteria and occurs independently of TLR4 signaling. Dis Model Mech 2017; 10:727-736. [PMID: 28450472 PMCID: PMC5483007 DOI: 10.1242/dmm.028589] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Accepted: 04/07/2017] [Indexed: 12/18/2022] Open
Abstract
Necrotizing enterocolitis (NEC) remains a leading cause of morbidity and mortality in premature infants. Both human surgical specimens and animal models suggest a potential involvement of Paneth cells in NEC pathogenesis. Paneth cells play critical roles in epithelial homeostasis, innate immunity and host-microbial interactions. Yet, the complex interplay between Paneth cell disruption, epithelial barrier dysfunction and microbial-driven inflammation remains unclear in the immature intestine. In this study, mucosal intestinal injury consistent with human NEC was induced in postnatal day 14-16 (P14-P16) mice by disrupting Paneth cells, followed by gavage with Klebsiella pneumonia. Mucosal injury was determined by histology, serum cytokine levels and epithelial barrier dysfunction. Toll-like receptor 4 (TLR4) activation was examined using protein expression, gene expression, and TLR4−/− mice. Finally, the role of bacteria was evaluated using heat-killed bacteria, conditioned media, Bacillus cereus and cecal slurries. We found that live bacteria were required to induce injury; however, TLR4 activation was not required. NEC induced by Paneth cell disruption results in altered localization of tight junction proteins and subsequent loss of barrier function. Prior research has shown a requirement for TLR4 activation to induce NEC-like damage. However, many infants develop NEC in the absence of Gram-negative rod bacteremia, raising the possibility that alternative pathways to intestinal injury exist. In this study, we show a previously unknown mechanism for the development of intestinal injury equivalent to that seen in human NEC and that is not dependent on TLR4 pathways. These data are congruent with the new hypothesis that NEC may be the consequence of several disease processes ending in a final common inflammatory pathway. Summary: Paneth cell disruption and bacterial exposure in immature murine intestine induces NEC-like pathology. Importantly, this occurs independently of TLR4 activation, presenting an alternative mechanism for disease development.
Collapse
Affiliation(s)
| | - Huiyu Gong
- Pediatrics, University of Iowa, Iowa City, IA 54424, USA
| | - Brock Pope
- Pediatrics, University of Iowa, Iowa City, IA 54424, USA
| | | | | |
Collapse
|
17
|
Poitout-Belissent F, Aulbach A, Tripathi N, Ramaiah L. Reducing blood volume requirements for clinical pathology testing in toxicologic studies-points to consider. Vet Clin Pathol 2016; 45:534-551. [DOI: 10.1111/vcp.12429] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|