1
|
Hegde M, Raj S, Pattanshetti AS, Nyamagoud SB. Gaining insights into chronic obstructive pulmonary disease exacerbation through emerging biomarkers and the chronic obstructive pulmonary disease assessment test score. Monaldi Arch Chest Dis 2024. [PMID: 38497202 DOI: 10.4081/monaldi.2024.2955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 02/26/2024] [Indexed: 03/19/2024] Open
Abstract
Chronic obstructive pulmonary disease (COPD), a leading cause of mortality and morbidity, presents significant challenges, particularly with exacerbations, which drastically impact patients' health and healthcare costs. The Global Initiative for Chronic Obstructive Lung Disease guidelines recommend comprehensive assessments beyond spirometry, with the COPD assessment test (CAT) emerging as a pivotal tool. Despite its utility, the relationship between CAT scores and specific biomarkers during exacerbations remains unclear. Hence, this study aims to assess the correlation between the CAT score and specific circulating biomarkers. A cross-sectional study from August 2023 to January 2024 included 59 COPD patients with exacerbations who underwent pulmonary function tests and completed the CAT score assessment. The CAT score cut-off point was set at 20, where a CAT score <20 indicated a low impact on health status and a CAT score ≥20 indicated a high impact on health status. On the same day, measurements of neutrophils, leukocytes, eosinophils, C-reactive protein, and procalcitonin were conducted. Patients with CAT scores ≥20 had significantly higher levels of neutrophils (p=0.001), leukocytes (p=0.006), procalcitonin (p=0.010), and forced expiratory volume in the first second/forced vital capacity (p=0.002), but lower eosinophil levels (p=0.025). A positive correlation existed between total CAT score and neutrophils (p=0.001), leukocytes (p=0.000), and procalcitonin (p=0.010), while eosinophil levels showed a negative correlation (p=0.025). The spirometry parameters showed no correlation with the total CAT score. This study highlights the link between CAT and key inflammatory biomarkers, supporting the use of blood biomarkers to identify COPD patients at risk of exacerbations.
Collapse
Affiliation(s)
- Megha Hegde
- Department of Pharmacy Practice, KLE College of Pharmacy, Hubli, Karnataka.
| | - Saurav Raj
- Department of Pharmacy Practice, KLE College of Pharmacy, Hubli, Karnataka.
| | | | | |
Collapse
|
2
|
Park HY, Yu JH. X-ray radiation-induced intestinal barrier dysfunction in human epithelial Caco-2 cell monolayers. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 264:115404. [PMID: 37625335 DOI: 10.1016/j.ecoenv.2023.115404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 08/18/2023] [Accepted: 08/21/2023] [Indexed: 08/27/2023]
Abstract
Radiation therapy and unwanted radiological or nuclear exposure, such as nuclear plant accidents, terrorist attacks, and military conflicts, pose serious health issues to humans. Dysfunction of the intestinal epithelial barrier and the leakage of luminal antigens and bacteria across the barrier have been linked to various human diseases. Intestinal permeability is regulated by intercellular structures, termed tight junctions (TJs), which are disrupted after radiation exposure. In this study, we investigated radiation-induced alterations in TJ-related proteins in an intestinal epithelial cell model. Caco-2 cells were irradiated with 2, 5, and 10 Gy and harvested 1 and 24 h after X-ray exposure. The trypan blue assay revealed that cell viability was reduced in a dose-dependent manner 24 h after X-ray exposure compared to that of non-irradiated cells. However, the WST-8 assay revealed that cell proliferation was significantly reduced only 24 h after radiation exposure to 10 Gy compared to that of non-irradiated cells. In addition, a decreased growth rate and increased doubling time were observed in cells irradiated with X-rays. Intestinal permeability was significantly increased, and transepithelial electrical resistance values were remarkably reduced in Caco-2 cell monolayers irradiated with X-rays compared to non-irradiated cells. X-ray irradiation significantly decreased the mRNA and protein levels of ZO-1, occludin, claudin-3, and claudin-4, with ZO-1 and claudin-3 protein levels decreasing in a dose-dependent manner. Overall, the present study reveals that exposure to X-ray induces dysfunction of the human epithelial intestinal barrier and integrity via the downregulation of TJ-related genes, which may be a key factor contributing to intestinal barrier damage and increased intestinal permeability.
Collapse
Affiliation(s)
- Ha-Young Park
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup 56212, Republic of Korea.
| | - Jin-Hee Yu
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup 56212, Republic of Korea
| |
Collapse
|
3
|
Pre-Administration of PLX-R18 Cells Protects Mice from Radiation-Induced Hematopoietic Failure and Lethality. Genes (Basel) 2022; 13:genes13101756. [PMID: 36292639 PMCID: PMC9601513 DOI: 10.3390/genes13101756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 09/22/2022] [Accepted: 09/23/2022] [Indexed: 11/23/2022] Open
Abstract
Acute Radiation Syndrome (ARS) is a syndrome involving damage to multiple organs caused by exposure to a high dose of ionizing radiation over a short period of time; even low doses of radiation damage the radiosensitive hematopoietic system and causes H-ARS. PLacenta eXpanded (PLX)-R18 is a 3D-expanded placenta-derived stromal cell product designated for the treatment of hematological disorders. These cells have been shown in vitro to secrete hematopoietic proteins, to stimulate colony formation, and to induce bone marrow migration. Previous studies in mice showed that PLX-R18 cells responded to radiation-induced hematopoietic failure by transiently secreting hematopoiesis related proteins to enhance reconstitution of the hematopoietic system. We assessed the potential effect of prophylactic PLX-R18 treatment on H-ARS. PLX-R18 cells were administered intramuscularly to C57BL/6 mice, −1 and 3 days after (LD70/30) total body irradiation. PLX R18 treatment significantly increased survival after irradiation (p < 0.0005). In addition, peripheral blood and bone marrow (BM) cellularity were monitored at several time points up to 30 days. PLX-R18 treatment significantly increased the number of colony-forming hematopoietic progenitors in the femoral BM and significantly raised peripheral blood cellularity. PLX-R18 administration attenuated biomarkers of bone marrow aplasia (EPO, FLT3L), sepsis (SAA), and systemic inflammation (sP-selectin and E-selectin) and attenuated radiation-induced inflammatory cytokines/chemokines and growth factors, including G-CSF, MIP-1a, MIP-1b, IL-2, IL-6 and MCP-1, In addition, PLX-R18 also ameliorated radiation-induced upregulation of pAKT. Taken together, prophylactic PLX-R18 administration may serve as a protection measure, mitigating bone marrow failure symptoms and systemic inflammation in the H-ARS model.
Collapse
|
4
|
Garg S, Garg TK, Miousse IR, Wise SY, Fatanmi OO, Savenka AV, Basnakian AG, Singh VK, Hauer-Jensen M. Effects of Gamma-Tocotrienol on Partial-Body Irradiation-Induced Intestinal Injury in a Nonhuman Primate Model. Antioxidants (Basel) 2022; 11:1895. [PMID: 36290618 PMCID: PMC9598988 DOI: 10.3390/antiox11101895] [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: 08/16/2022] [Revised: 09/16/2022] [Accepted: 09/19/2022] [Indexed: 11/17/2022] Open
Abstract
Exposure to high doses of radiation, accidental or therapeutic, often results in gastrointestinal (GI) injury. To date, there are no therapies available to mitigate GI injury after radiation exposure. Gamma-tocotrienol (GT3) is a promising radioprotector under investigation in nonhuman primates (NHP). We have shown that GT3 has radioprotective function in intestinal epithelial and crypt cells in NHPs exposed to 12 Gy total-body irradiation (TBI). Here, we determined GT3 potential in accelerating the GI recovery in partial-body irradiated (PBI) NHPs using X-rays, sparing 5% bone marrow. Sixteen rhesus macaques were treated with either vehicle or GT3 24 h prior to 12 Gy PBI. Structural injuries and crypt survival were examined in proximal jejunum on days 4 and 7. Plasma citrulline was assessed using liquid chromatography-tandem mass spectrometry (LC-MS/MS). Crypt cell proliferation and apoptotic cell death were evaluated using Ki-67 and TUNEL staining. PBI significantly decreased mucosal surface area and reduced villous height. Interestingly, GT3 increased crypt survival and enhanced stem cell proliferation at day 4; however, the effects seemed to be minimized by day 7. GT3 did not ameliorate a radiation-induced decrease in citrulline levels. These data suggest that X-rays induce severe intestinal injury post-PBI and that GT3 has minimal radioprotective effect in this novel model.
Collapse
Affiliation(s)
- Sarita Garg
- Division of Radiation Health, Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Tarun K. Garg
- UAMS Myeloma Center, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Isabelle R. Miousse
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Stephen Y. Wise
- Division of Radioprotectants, Department of Pharmacology and Molecular Therapeutics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Oluseyi O. Fatanmi
- Division of Radioprotectants, Department of Pharmacology and Molecular Therapeutics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Alena V. Savenka
- Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Alexei G. Basnakian
- Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- John L. McClellan Memorial VA Hospital, Central Arkansas Veterans Healthcare System, Little Rock, AR 72205, USA
| | - Vijay K. Singh
- Division of Radioprotectants, Department of Pharmacology and Molecular Therapeutics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Martin Hauer-Jensen
- Division of Radiation Health, Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| |
Collapse
|
5
|
Winters TA, Taliaferro LP, Satyamitra MM. Development of Biomarkers for Radiation Biodosimetry and Medical Countermeasures Research: Current Status, Utility, and Regulatory Pathways. Radiat Res 2022; 197:554-558. [DOI: 10.1667/rade-21-00213.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 01/05/2022] [Indexed: 11/03/2022]
Affiliation(s)
- Thomas A. Winters
- Radiation and Nuclear Countermeasures Program (RNCP), Division of Allergy, Immunology and Transplantation (DAIT), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, Maryland
| | - Lanyn P. Taliaferro
- Radiation and Nuclear Countermeasures Program (RNCP), Division of Allergy, Immunology and Transplantation (DAIT), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, Maryland
| | - Merriline M. Satyamitra
- Radiation and Nuclear Countermeasures Program (RNCP), Division of Allergy, Immunology and Transplantation (DAIT), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, Maryland
| |
Collapse
|
6
|
Ewing LE, Biju PG, Pathak R, Melnyk S, Hauer-Jensen M, Koturbash I. Methods for induction and assessment of intestinal permeability in rodent models of radiation injury. Methods Cell Biol 2022; 168:235-247. [PMID: 35366985 PMCID: PMC9808921 DOI: 10.1016/bs.mcb.2021.12.017] [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] [Indexed: 01/14/2023]
Abstract
Ionizing radiation (IR) is a significant contributor to the contemporary market of energy production and an important diagnostic and treatment modality. Besides having numerous useful applications, it is also a ubiquitous environmental stressor and a potent genotoxic and epigenotoxic agent, capable of causing substantial damage to organs and tissues of living organisms. The gastrointestinal (GI) tract is highly sensitive to IR. This problem is further compounded by the fact that there is no FDA-approved medication to mitigate acute radiation-induced GI syndrome. Therefore, establishing the animal model for studying IR-induced GI-injury is crucially important to understand the harmful consequences of intestinal radiation damage. Here, we discuss two different animal models of IR-induced acute gastrointestinal syndrome and two separate methods for measuring the magnitude of intestinal radiation damage.
Collapse
Affiliation(s)
- Laura E Ewing
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Prabath G Biju
- Department of Biochemistry, University of Kerala, Trivandrum, India
| | - Rupak Pathak
- Division of Radiation Health, Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Stepan Melnyk
- Arkansas Children's Research Institute, Little Rock, AR, United States
| | - Martin Hauer-Jensen
- Division of Radiation Health, Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Igor Koturbash
- Department of Environmental and Occupational Health, College of Public Health, University of Arkansas for Medical Sciences, Little Rock, AR, United States; Center for Dietary Supplements Research, University of Arkansas for Medical Sciences, Little Rock, AR, United States.
| |
Collapse
|
7
|
Satyamitra MM, DiCarlo AL, Hollingsworth BA, Winters TA, Taliaferro LP. Development of Biomarkers for Radiation Biodosimetry and Medical Countermeasures Research: Current Status, Utility, and Regulatory Pathways. Radiat Res 2021; 197:514-532. [PMID: 34879151 DOI: 10.1667/rade-21-00157.1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 10/22/2021] [Indexed: 11/03/2022]
Abstract
Biomarkers are important indicators of biological processes in health or disease. For this reason, they play a critical role in advanced development of radiation biodosimetry tools and medical countermeasures (MCMs). They can aid in the assessment of radiation exposure level, extent of radiation-induced injury, and/or efficacy of an MCM. This meeting report summarizes the presentations and discussions from the 2020 workshop titled, "Biomarkers in Radiation Biodosimetry and Medical Countermeasures," sponsored by the Radiation and Nuclear Countermeasures Program (RNCP) at the National Institute of Allergy and Infectious Diseases (NIAID). The main goals of this meeting were to: 1. Provide an overview on biomarkers and to focus on the state of science with regards to biomarkers specific to radiation biodosimetry and MCMs; 2. Understand developmental challenges unique to the role of biomarkers in the fields of radiation biodosimetry and MCM development; and 3. Identify existing gaps and needs for translational application.
Collapse
Affiliation(s)
- Merriline M Satyamitra
- Radiation and Nuclear Countermeasures Program (RNCP), Division of Allergy, Immunology and Transplantation (DAIT), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, Maryland
| | - Andrea L DiCarlo
- Radiation and Nuclear Countermeasures Program (RNCP), Division of Allergy, Immunology and Transplantation (DAIT), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, Maryland
| | - Brynn A Hollingsworth
- Radiation and Nuclear Countermeasures Program (RNCP), Division of Allergy, Immunology and Transplantation (DAIT), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, Maryland
| | - Thomas A Winters
- Radiation and Nuclear Countermeasures Program (RNCP), Division of Allergy, Immunology and Transplantation (DAIT), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, Maryland
| | - Lanyn P Taliaferro
- Radiation and Nuclear Countermeasures Program (RNCP), Division of Allergy, Immunology and Transplantation (DAIT), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, Maryland
| |
Collapse
|
8
|
Fang Z, Chen P, Tang S, Chen A, Zhang C, Peng G, Li M, Chen X. Will mesenchymal stem cells be future directions for treating radiation-induced skin injury? Stem Cell Res Ther 2021; 12:179. [PMID: 33712078 PMCID: PMC7952822 DOI: 10.1186/s13287-021-02261-5] [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: 01/20/2021] [Accepted: 03/01/2021] [Indexed: 01/09/2023] Open
Abstract
Radiation-induced skin injury (RISI) is one of the common serious side effects of radiotherapy (RT) for patients with malignant tumors. Mesenchymal stem cells (MSCs) are applied to RISI repair in some clinical cases series except some traditional options. Though direct replacement of damaged cells may be achieved through differentiation capacity of MSCs, more recent data indicate that various cytokines and chemokines secreted by MSCs are involved in synergetic therapy of RISI by anti-inflammatory, immunomodulation, antioxidant, revascularization, and anti-apoptotic activity. In this paper, we not only discussed different sources of MSCs on the treatment of RISI both in preclinical studies and clinical trials, but also summarized the applications and mechanisms of MSCs in other related regenerative fields.
Collapse
Affiliation(s)
- Zhuoqun Fang
- Department of Plastic Surgery, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, 350001, Fujian, People's Republic of China
| | - Penghong Chen
- Department of Plastic Surgery, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, 350001, Fujian, People's Republic of China
| | - Shijie Tang
- Department of Plastic Surgery, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, 350001, Fujian, People's Republic of China
| | - Aizhen Chen
- Department of Plastic Surgery, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, 350001, Fujian, People's Republic of China
| | - Chaoyu Zhang
- Department of Plastic Surgery, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, 350001, Fujian, People's Republic of China
| | - Guohao Peng
- Department of Plastic Surgery, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, 350001, Fujian, People's Republic of China
| | - Ming Li
- Department of Plastic Surgery, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, 350001, Fujian, People's Republic of China
| | - Xiaosong Chen
- Department of Plastic Surgery, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, 350001, Fujian, People's Republic of China.
| |
Collapse
|
9
|
Patterson AM, Plett PA, Chua HL, Sampson CH, Fisher A, Feng H, Unthank JL, Miller SJ, Katz BP, MacVittie TJ, Orschell CM. Development of a Model of the Acute and Delayed Effects of High Dose Radiation Exposure in Jackson Diversity Outbred Mice; Comparison to Inbred C57BL/6 Mice. HEALTH PHYSICS 2020; 119:633-646. [PMID: 32932286 PMCID: PMC9374540 DOI: 10.1097/hp.0000000000001344] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
Development of medical countermeasures against radiation relies on robust animal models for efficacy testing. Mouse models have advantages over larger species due to economics, ease of conducting aging studies, existence of historical databases, and research tools allowing for sophisticated mechanistic studies. However, the radiation dose-response relationship of inbred strains is inherently steep and sensitive to experimental variables, and inbred models have been criticized for lacking genetic diversity. Jackson Diversity Outbred (JDO) mice are the most genetically diverse strain available, developed by the Collaborative Cross Consortium using eight founder strains, and may represent a more accurate model of humans than inbred strains. Herein, models of the Hematopoietic-Acute Radiation Syndrome and the Delayed Effects of Acute Radiation Exposure were developed in JDO mice and compared to inbred C57BL/6. The dose response relationship curve in JDO mice mirrored the more shallow curves of primates and humans, characteristic of genetic diversity. JDO mice were more radioresistant than C57BL/6 and differed in sensitivity to antibiotic countermeasures. The model was validated with pegylated-G-CSF, which provided significantly enhanced 30-d survival and accelerated blood recovery. Long-term JDO survivors exhibited increased recovery of blood cells and functional bone marrow hematopoietic progenitors compared to C57BL/6. While JDO hematopoietic stem cells declined more in number, they maintained a greater degree of quiescence compared to C57BL/6, which is essential for maintaining function. These JDO radiation models offer many of the advantages of small animals with the genetic diversity of large animals, providing an attractive alternative to currently available radiation animal models.
Collapse
Affiliation(s)
- Andrea M. Patterson
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN
| | - P. Artur Plett
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN
| | - Hui Lin Chua
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN
| | - Carol H. Sampson
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN
| | - Alexa Fisher
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN
| | - Hailin Feng
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN
| | - Joseph L. Unthank
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN
| | - Steve J. Miller
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN
| | - Barry P. Katz
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN
| | - Thomas J. MacVittie
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD
| | | |
Collapse
|
10
|
Reevaluation of Lung Injury in TNF-Induced Shock: The Role of the Acid Sphingomyelinase. Mediators Inflamm 2020; 2020:3650508. [PMID: 32410851 PMCID: PMC7211256 DOI: 10.1155/2020/3650508] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 02/14/2020] [Accepted: 02/27/2020] [Indexed: 01/06/2023] Open
Abstract
Tumor necrosis factor (TNF) is a well-known mediator of sepsis. In many cases, sepsis results in multiple organ injury including the lung with acute respiratory distress syndrome (ARDS). More than 20-year-old studies have suggested that TNF may be directly responsible for organ injury during sepsis. However, these old studies are inconclusive, because they relied on human rather than conspecific TNF, which was contaminated with endotoxin in most studies. In this study, we characterized the direct effects of intravenous murine endotoxin-free TNF on cardiovascular functions and organ injury in mice with a particular focus on the lungs. Because of the relevance of the acid sphingomyelinase in sepsis, ARDS, and caspase-independent cell death, we also included acid sphingomyelinase-deficient (ASM−/−) mice. ASM−/− and wild-type (WT) mice received 50 μg endotoxin-free murine TNF intravenously alone or in combination with the pan-caspase inhibitor carbobenzoxy-valyl-alanyl-aspartyl-[O-methyl]-fluoromethylketone (zVAD) and were ventilated at low tidal volume while lung mechanics were followed. Blood pressure was stabilized by intra-arterial fluid support, and body temperature was kept at 37°C to delay lethal shock and to allow investigation of blood gases, lung histopathology, proinflammatory mediators, and microvascular permeability 6 hours after TNF application. Besides the lungs, also the kidneys and liver were examined. TNF elicited the release of inflammatory mediators and a high mortality rate, but failed to injure the lungs, kidneys, or liver of healthy mice significantly within 6 hours. Mortality in WT mice was most likely due to sepsis-like shock, as indicated by metabolic acidosis, high procalcitonin levels, and cardiovascular failure. ASM−/− mice were protected from TNF-induced hypotension and reflex tachycardia and also from mortality. In WT mice, intravenous exogenous TNF does not cause organ injury but induces a systemic inflammatory response with cardiovascular failure, in which the ASM plays a role.
Collapse
|
11
|
Gupta R, Yin L, Grosche A, Lin S, Xu X, Guo J, Vaught LA, Okunieff PG, Vidyasagar S. An Amino Acid-Based Oral Rehydration Solution Regulates Radiation-Induced Intestinal Barrier Disruption in Mice. J Nutr 2020; 150:1100-1108. [PMID: 32133527 DOI: 10.1093/jn/nxaa025] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 01/06/2020] [Accepted: 01/28/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Radiotherapy inadvertently affects gastrointestinal (GI) epithelial cells, causing intestinal barrier disruption and increased permeability. OBJECTIVE We examined the effect of amino acid-based oral rehydration solution (AA-ORS) on radiation-induced changes of intestinal barrier function and epithelial tight junctions (TJs) in a randomized experimental study using a total-body irradiation (TBI) mouse model. METHODS Eight-week-old male Swiss mice received a single-dose TBI (0, 1, 3, or 5 Gy), and subsequent gastric gavage with AA-ORS (threonine, valine, serine, tyrosine, and aspartic acid) or saline for 2 or 6 d. Intestinal barrier function of mouse ileum was characterized by electrophysiological analysis of conductance, anion selectivity, and paracellular permeability [fluorescein isothiocyanate (FITC)-dextran]. Ultrastructural changes of TJs were evaluated by transmission electron microscopy. Membrane protein and mRNA expression of claudin-1, -2, -3, -5, and -7, occludin, and E-cadherin were analyzed with western blot, qPCR, and immunohistochemistry. Nonparametric tests were used to compare treatment-dose differences for each time point. RESULTS Saline-treated mice had a higher conductance at doses as low as 3 Gy, and as early as 2 d post-TBI compared with 0 Gy (P < 0.001). Paracellular permeability and dilution potential were increased 6 d after 5 Gy TBI (P < 0.001). Conductance decreased with AA-ORS after 2 d in 3-Gy and 5-Gy mice (P < 0.05 and P < 0.001), and on day 6 after 5 Gy TBI (P < 0.001). Anion selectivity and FITC permeability decreased from 0.73 ± 0.02 to 0.61 ± 0.03 pCl/pNa (P < 0.01) and from 2.7 ± 0.1 × 105 to 2.1 ± 0.1 × 105 RFU (P < 0.001) in 5-Gy mice treated with AA-ORS for 6 d compared with saline. Irradiation-induced ultrastructural changes of TJs characterized by decreased electron density and gap formation improved with AA-ORS. Reduced claudin-1, -3, and -7 membrane expression after TBI recovered with AA-ORS within 6 d, whereas claudin-2 decreased indicating restitution of TJ proteins. CONCLUSIONS Radiation-induced functional and structural disruption of the intestinal barrier in mice is reversed by AA-ORS rendering AA-ORS a potential treatment option in prospective clinical trials in patients with gastrointestinal barrier dysfunction.
Collapse
Affiliation(s)
- Reshu Gupta
- Entrinsic Health Solutions, Norwood, MA, USA
| | - Liangjie Yin
- Department of Radiation Oncology, University of Florida Shands Cancer Center, Gainesville, FL, USA
| | | | | | - Xiaodong Xu
- Department of Radiation Oncology, University of Florida Shands Cancer Center, Gainesville, FL, USA
| | - Jing Guo
- Department of Radiation Oncology, University of Florida Shands Cancer Center, Gainesville, FL, USA
| | - Lauren A Vaught
- Department of Radiation Oncology, University of Florida Shands Cancer Center, Gainesville, FL, USA
| | - Paul G Okunieff
- Department of Radiation Oncology, University of Florida Shands Cancer Center, Gainesville, FL, USA
| | - Sadasivan Vidyasagar
- Department of Radiation Oncology, University of Florida Shands Cancer Center, Gainesville, FL, USA
| |
Collapse
|
12
|
Kim DW, An JM, Hong SP, Hahm KB. Predictive Value of Procalcitonin in Acute Severe Ulcerative Colitis: Not Quite PerfeCT? Dig Dis Sci 2019; 64:3033-3034. [PMID: 30734240 DOI: 10.1007/s10620-019-05481-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
Affiliation(s)
- Deok Whan Kim
- Digestive Disease Center, CHA University Bundang Medical Center, 59 Yatap-ro, Bundang-gu, Seongnam, Korea
| | - Jeong Min An
- Digestive Disease Center, CHA University Bundang Medical Center, 59 Yatap-ro, Bundang-gu, Seongnam, Korea
| | - Seong Pyo Hong
- Digestive Disease Center, CHA University Bundang Medical Center, 59 Yatap-ro, Bundang-gu, Seongnam, Korea
| | - Ki Baik Hahm
- Digestive Disease Center, CHA University Bundang Medical Center, 59 Yatap-ro, Bundang-gu, Seongnam, Korea.
| |
Collapse
|
13
|
Serum Procalcitonin as a Potential Early Predictor of Short-Term Outcomes in Acute Severe Ulcerative Colitis. Dig Dis Sci 2019; 64:3263-3273. [PMID: 30604378 DOI: 10.1007/s10620-018-5446-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2018] [Accepted: 12/20/2018] [Indexed: 12/13/2022]
Abstract
BACKGROUND Delayed colectomy can be life-threatening for patients with acute severe ulcerative colitis (ASUC). However, few biomarkers can predict the outcomes of ASUC patients before treatment. Serum procalcitonin (PCT) has been observed to be increased in ASUC patients. AIM The aim of this study was to estimate the association between serum PCT and short-term outcomes in patients with ASUC. METHODS A single-center observational study was conducted at a referral hospital from January 2012 to January 2018. Hospitalized ASUC patients, who were administered intravenous corticosteroids (IVCS), were enrolled and followed up for 6 months. The primary outcome was IVCS failure; the secondary outcome was colectomy. Relationships between indicators and clinical outcomes were assessed. RESULTS Of 152 ASUC patients enrolled in this study, 81 responded to IVCS and 71 failed (62 required short-term colectomy and 9 responded to second-line rescue therapy). Serum PCT on admission was significantly higher in IVCS-failure cases and surgical cases than in medical responders. Serum PCT ≥ 0.10 µg/L (OR = 4.134, p = 0.001) predicted IVCS failure with specificity of 0.741, and the combined measurement with fecal calprotectin (FC) ≥ 1500 µg/g improved the sensitivity. Serum PCT correlated significantly with the Ulcerative Colitis Endoscopic Index of Severity (r = 0.416, p < 0.001) and FC (r = 0.384, p < 0.001). CONCLUSION Serum PCT on admission could be a potential early non-invasive predictive biomarker for IVCS failure in ASUC patients, and a combination of PCT and FC could improve the predictive value.
Collapse
|
14
|
Interaction between high-fat diet and ethanol intake leads to changes on the fecal microbiome. J Nutr Biochem 2019; 72:108215. [DOI: 10.1016/j.jnutbio.2019.07.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 06/24/2019] [Accepted: 07/19/2019] [Indexed: 02/07/2023]
|
15
|
C/EBPδ protects from radiation-induced intestinal injury and sepsis by suppression of inflammatory and nitrosative stress. Sci Rep 2019; 9:13953. [PMID: 31562350 PMCID: PMC6764943 DOI: 10.1038/s41598-019-49437-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 08/15/2019] [Indexed: 12/20/2022] Open
Abstract
Ionizing radiation (IR)-induced intestinal damage is characterized by a loss of intestinal crypt cells, intestinal barrier disruption and translocation of intestinal microflora resulting in sepsis-mediated lethality. We have shown that mice lacking C/EBPδ display IR-induced intestinal and hematopoietic injury and lethality. The purpose of this study was to investigate whether increased IR-induced inflammatory, oxidative and nitrosative stress promote intestinal injury and sepsis-mediated lethality in Cebpd−/− mice. We found that irradiated Cebpd−/− mice show decreased villous height, crypt depth, crypt to villi ratio and expression of the proliferation marker, proliferating cell nuclear antigen, indicative of intestinal injury. Cebpd−/− mice show increased expression of the pro-inflammatory cytokines (Il-6, Tnf-α) and chemokines (Cxcl1, Mcp-1, Mif-1α) and Nos2 in the intestinal tissues compared to Cebpd+/+ mice after exposure to TBI. Cebpd−/− mice show decreased GSH/GSSG ratio, increased S-nitrosoglutathione and 3-nitrotyrosine in the intestine indicative of basal oxidative and nitrosative stress, which was exacerbated by IR. Irradiated Cebpd-deficient mice showed upregulation of Claudin-2 that correlated with increased intestinal permeability, presence of plasma endotoxin and bacterial translocation to the liver. Overall these results uncover a novel role for C/EBPδ in protection against IR-induced intestinal injury by suppressing inflammation and nitrosative stress and underlying sepsis-induced lethality.
Collapse
|
16
|
The pathogenesis of mucositis: updated perspectives and emerging targets. Support Care Cancer 2019; 27:4023-4033. [PMID: 31286231 DOI: 10.1007/s00520-019-04893-z] [Citation(s) in RCA: 109] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 05/22/2019] [Indexed: 12/13/2022]
Abstract
Mucositis research and treatment are a rapidly evolving field providing constant new avenues of research and potential therapies. The MASCC/ISOO Mucositis Study Group regularly assesses available literature relating to pathogenesis, mechanisms, and novel therapeutic approaches and distils this to summary perspectives and recommendations. Reviewers assessed 164 articles published between January 2011 and June 2016 to identify progress made since the last review and highlight new targets for further investigation. Findings were organized into sections including established and emerging mediators of toxicity, potential insights from technological advances in mucositis research, and perspective. Research momentum is accelerating for mucositis pathogenesis, and with this has come utilization of new models and interventions that target specific mechanisms of injury. Technological advances have the potential to revolutionize the field of mucositis research, although focused effort is needed to move rationally targeted interventions to the clinical setting.
Collapse
|
17
|
Kaur A, Ten Have GAM, Hritzo B, Deutz NEP, Olsen C, Moroni M. Morphological and functional impairment in the gut in a partial body irradiation minipig model of GI-ARS. Int J Radiat Biol 2019; 96:112-128. [PMID: 30475652 DOI: 10.1080/09553002.2018.1552377] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Purpose: Göttingen minipig (G-MP) displays classic gastrointestinal acute radiation syndrome (GI-ARS) following total body irradiation (TBI) at GI doses which are lethal by 10-14 days. In collaboration with BARDA, we are developing a hemi-body/partial body irradiation (PBI) model by exposing only the abdomen and lower extremities to study GI structure/function impairment, natural history of injury and recovery, as well as correlative biomarkers out to 30 days.Materials and methods: Twenty-four G-MP were exposed to either 12 or 16 Gy (LINAC Elekta); head, forelimbs, and thorax were outside the irradiation field, sparing ∼50% of the bone marrow. Animals were followed for 30 days with euthanasia scheduled at pre-set intervals to study the time course of GI injury and recovery. Hematological profiles, clinical symptoms, gross- and histo-pathology including markers of proliferation and apoptosis in the small intestines, gut function parameters (food tolerance, digestion, absorption, citrulline production), and levels of two biomarkers, CRP and IGF-1, were evaluated.Results: PBI at 16 Gy yielded higher lethality than 12 Gy. Unlike TBI, PBI did not cause severe pancytopenia or external hemorrhage, as expected, and allowed to focus the injury on GI organs while sparing the radiation sensitive heart and lung. Compromised animals showed inactivity, anorexia, vomiting, diarrhea, and weight loss. Histology revealed that in 12 Gy irradiated animals, lesions recovered overtime. In 16 Gy irradiated animals, lesions were more pronounced and persistent. BrdU and Ki67 labelling demonstrated dose-dependent loss of crypts and subsequent mucosal ulceration which recovered over time. Minimal apoptosis was observed at both doses. Reductions in food tolerance, digestion, absorption, and citrulline production were time and dose-dependent. Loss of citrulline reached a nadir between 6-12 days and then recovered partially. CRP and IGF-1 were upregulated following PBI at GI doses.Conclusions: This lower hemi-body irradiation model allowed for extended survival at GI-specific ARS doses and development of a well-controlled GI syndrome with minimal hematopoietic injury or confounding mortality from cardiopulmonary damage. A dose-dependent impairment in the intestinal structure resulted in overall decreased gut functionality followed by a partial recovery. However, while the structure appeared to be recovered, not all functionality was attained. PBI induced systemic inflammation and altered the IGF-1 hormone indicating that these can be used as biomarkers in the minipig even under partial body conditions. This PBI model aligns with other minipig models under BARDA's large animal consortium to test medical countermeasure efficacy against a less complex GI-specific ARS injury.
Collapse
Affiliation(s)
- Amandeep Kaur
- Radiation Countermeasure program, Scientific Research Department, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Gabriëlla A M Ten Have
- Center for Translational Research in Aging & Longevity, Department of Health & Kinesiology, Texas A&M University, College Station, TX, USA
| | - Bernadette Hritzo
- Radiation Countermeasure program, Scientific Research Department, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Nicolaas E P Deutz
- Center for Translational Research in Aging & Longevity, Department of Health & Kinesiology, Texas A&M University, College Station, TX, USA
| | - Cara Olsen
- Biostatistics Consulting Center, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Maria Moroni
- Radiation Countermeasure program, Scientific Research Department, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| |
Collapse
|
18
|
Ossetrova NI, Stanton P, Krasnopolsky K, Ismail M, Doreswamy A, Hieber KP. Biomarkers for Radiation Biodosimetry and Injury Assessment after Mixed-field (Neutron and Gamma) Radiation in the Mouse Total-body Irradiation Model. HEALTH PHYSICS 2018; 115:727-742. [PMID: 30299338 DOI: 10.1097/hp.0000000000000938] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
The risk of potential radiation exposure scenarios that include detonation of nuclear weapons, terrorist attacks on nuclear reactors, and the use of conventional explosives to disperse radioactive substances has increased in recent years. The majority of radiation biodosimetry and countermeasure studies have been performed using photon radiation even though many exposure scenarios predict mixed-field (neutron and photon) radiation. Hence, there is a need to evaluate biomarkers and accurately determine exposure levels of mixed-field combinations of neutrons and photons for an individual. These biomarkers will be critical for biodosimetry triage, treatment, and follow-up visits with such individuals. We evaluated the utility of multiple blood biomarkers for early response assessment of radiation exposure using a mouse (B6D2F1, males and females) total-body irradiation model exposed to a mixed-field (neutrons and gamma rays) using the Armed Forces Radiobiology Research Institute's Mark F nuclear research reactor. Total-body irradiation was given as a single exposure over a dose range from 1.5 to 6 Gy, dose rates of 0.6 and 1.9 Gy min, and different proportions of neutrons and gammas: either (67% neutrons + 33% gammas) or (30% neutrons + 70% gammas). Blood was collected 1, 2, 4, and 7 d after total-body irradiation. Radiation-responsive protein biomarkers were measured using the Meso Scale Diagnostics' high-throughput MULTI-ARRAY plate-format platform (QuickPlex 120 Imager) and enzyme-linked immunosorbent assay kits. Results demonstrate (1) dose- and time-dependent changes in fms-related tyrosine kinase 3 ligand, interleukins IL-5, IL-10, IL-12, and IL-18, granulocyte and granulocyte-macrophage colony-stimulating factors, thrombopoietin, erythropoietin, acute-phase proteins (serum amyloid A and lipopolysaccharide binding protein), surface plasma neutrophil (CD45) and lymphocyte (CD27) markers, ratio of CD45 to CD27, and procalcitonin; (2) dose- and time-dependent changes in blood cell counts (lymphocytes, neutrophils, platelets, red blood cells, and ratio of neutrophils to lymphocytes); (3) levels of IL-18, granulocyte and granulocyte-macrophage colony-stimulating factors, serum amyloid A, and procalcitonin were significantly higher in animals irradiated with 67% neutrons + 33% gammas compared to those irradiated with 30% neutrons + 70% gammas (p < 0.015), while no significant differences (p > 0.114) were observed in hematological biomarker counts; (4) exposure with 3-fold difference in dose rate (0.6 or 1.9 Gy min) revealed no significant differences in hematological and protein biomarker levels (p > 0.154); and (5) no significant differences in hematological and protein biomarker levels were observed in the sex-comparison study for any radiation dose at any time after exposure (p > 0.088). Results show that the dynamic changes in the levels of selected hematopoietic cytokines, organ-specific biomarkers, and acute-phase protein biomarkers reflect the time course and severity of acute radiation syndrome and may function as prognostic indicators of acute radiation syndrome outcome. These studies supplement an ongoing effort to deliver U.S. Federal Drug Administration-approved biodosimetry capabilities, which assess mixed-field radiation exposure.
Collapse
Affiliation(s)
- Natalia I Ossetrova
- 1Uniformed Services University, Armed Forces Radiobiology Research Institute, Scientific Research Department, 4555 South Palmer Road, Bethesda, MD 20889-5648
| | | | | | | | | | | |
Collapse
|
19
|
Ossetrova NI, Stanton P, Krasnopolsky K, Ismail M, Doreswamy A, Hieber KP. Comparison of Biodosimetry Biomarkers for Radiation Dose and Injury Assessment After Mixed-Field (Neutron and Gamma) and Pure Gamma Radiation in the Mouse Total-Body Irradiation Model. HEALTH PHYSICS 2018; 115:743-759. [PMID: 33289997 DOI: 10.1097/hp.0000000000000939] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
The detonation of a nuclear weapon and the occurrence of a nuclear accident represent possible mass-casualty events with significant exposure to mixed neutron and gamma radiation fields in the first few minutes after the event with the ensuing fallout, extending for miles from the epicenter, that would result primarily in photon (gamma- and/or x-ray) exposure. Circulating biomarkers represent a crucial source of information in a mass-casualty radiation exposure triage scenario. We evaluated multiple blood biodosimetry and organ-specific biomarkers for early-response assessment of radiation exposure using a mouse (B6D2F1, males and females) total-body irradiation model exposed to Co gamma rays over a broad dose range (3-12 Gy) and dose rates of either 0.6 or 1.9 Gy min and compared the results with those obtained after exposure of mice to a mixed field (neutrons and gamma rays) using the Armed Forces Radiobiology Research Institute Co gamma-ray source and TRIGA Mark F nuclear research reactor. The mixed-field studies were performed previously over a broad dose range (1.5-6 Gy), with dose rates of either 0.6 or 1.9 Gy min, and using different proportions of neutrons and gammas: either (67% neutrons + 33% gammas) or (30% neutrons + 70% gammas). Blood was collected 1, 2, 4, and 7 d after total-body irradiation. Results from Co gamma-ray studies demonstrate: (1) significant dose- and time-dependent reductions in circulating mature hematopoietic cells; (2) dose- and time-dependent changes in fms-related tyrosine kinase 3 ligand, interleukins IL-5, IL-10, IL-12, and IL-18, granulocyte colony-stimulating factors, thrombopoietin, erythropoietin, acute-phase proteins (serum amyloid A and lipopolysaccharide binding protein), surface plasma neutrophil (CD45) and lymphocyte (CD27) markers, ratio of CD45 to CD27, procalcitonin but not in intestinal fatty acid binding protein; (3) no significant differences were observed between dose-rate groups in hematological and protein profiles (fms-related tyrosine kinase 3 ligand, IL-5, IL-12, IL-18, erythropoietin, granulocyte colony-stimulating factor, granulocyte-macrophage colony-stimulating factor, CD27, CD45, and ratio of CD45 to CD27) for any radiation dose at any time after exposure (p > 0.148); (4) no significant differences were observed between sex groups in hematological and protein profiles (fms-related tyrosine kinase 3 ligand, IL-18, erythropoietin, granulocyte colony-stimulating factor, granulocyte-macrophage colony-stimulating factor, serum amyloid A, CD45) for any radiation dose at any time after exposure (p > 0.114); and (5) PCT level significantly increased (p < 0.008) in mice irradiated with 12 Gy on day 7 post-total-body irradiation without significant differences between groups irradiated at dose rates of either 0.6 or 1.9 Gy min (p > 0.287). Radiation-quality comparison results demonstrate that: (1) equivalent doses of pure gamma rays and mixed-field radiation do not produce equivalent biological effects, and hematopoietic syndrome occurs at lower doses of mixed-field radiation; (2) ratios of hematological and protein biomarker means in the Co study compared to mixed-field studies using 2× Co doses vs. 1× TRIGA radiation doses (i.e., 3 Gy Co vs. 1.5 Gy TRIGA) ranged from roughly 0.2 to as high as 26.5 but 57% of all ratios fell within 0.7 and 1.3; and (3) in general, biomarker results are in agreement with the relative biological effectiveness = 1.95 (Dn/Dt = 0.67) reported earlier by Armed Forces Radiobiology Research Institute scientists in mouse survival countermeasure studies.
Collapse
Affiliation(s)
- Natalia I Ossetrova
- 1Uniformed Services University, Armed Forces Radiobiology Research Institute, Scientific Research Department, 4555 South Palmer Road Bethesda, MD 20889-5648
| | | | | | | | | | | |
Collapse
|
20
|
Xu MX, Ge CX, Qin YT, Gu TT, Lou DS, Li Q, Hu LF, Tan J. Multicombination Approach Suppresses Listeria monocytogenes-Induced Septicemia-Associated Acute Hepatic Failure: The Role of iRhom2 Signaling. Adv Healthc Mater 2018; 7:e1800427. [PMID: 29944201 DOI: 10.1002/adhm.201800427] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2018] [Revised: 05/26/2018] [Indexed: 12/18/2022]
Abstract
The mortality rate of acute liver failure significantly increases due to fatal septicemia. Inactive rhomboid protein 2 (iRhom2) is an essential regulator of shedding TNF-α by trafficking with TNF-α converting enzyme (TACE). Fisetin, a flavonoid present in various fruits and plants, possesses anti-oxidative stress and anti-inflammatory activities. Here, multi-combination nanoparticles Fe@Au conjugated with fisetin, iRhom2 small interfering RNA (siRNA), and TNF-α inhibitor (FN) are prepared to examine their effects on fatal septicemia-associated hepatic failure induced by Listeria monocytogenes (LM) in mice and to reveal the underlying mechanisms. After LM infection, upregulation of glutamic-oxalacetic transaminease, glutamic-pyruvic transaminase, alkaline phosphatase, TNF-α, malondialdehyde, H2 O2 , and O2- is observedcompared to FN-treated mice. The iRhom2/TACE/TNF-α signals are enhanced in vivo and in vitro, resulting in oxidative stress, which is especially associated with the activation of kupffer cells and other macrophages. Decrease in Nrf2 activation and increase of inflammation-associated regulators are also noted in vivo and in vitro. Furthermore, overexpression of TNF-α derived from macrophages aggravates hepatic failure. Inversely, the processes above are restored by FN nanoparticles through the regulation of the iRhom2/TACE/TNF-α axis and Nrf2 activation. These findings suggest that FN may be a potential approach to protect against bacterial septicemia-related diseases by targeting iRhom2.
Collapse
Affiliation(s)
- Min-Xuan Xu
- Chongqing Key Laboratory of Medicinal Resources in the Three Gorges Reservoir Region; School of Biological and Chemical Engineering; Chongqing University of Education; Chongqing 400067 P. R. China
- Research Center of Brain Intellectual Promotion and Development for Children Aged 0-6 Years; Chongqing University of Education; Chongqing 400067 P. R. China
| | - Chen-Xu Ge
- Chongqing Key Laboratory of Medicinal Resources in the Three Gorges Reservoir Region; School of Biological and Chemical Engineering; Chongqing University of Education; Chongqing 400067 P. R. China
- Research Center of Brain Intellectual Promotion and Development for Children Aged 0-6 Years; Chongqing University of Education; Chongqing 400067 P. R. China
| | - Yu-Ting Qin
- School of Medicine and Pharmacy; Ocean University of China; Qingdao 266100 P. R. China
| | - Ting-Ting Gu
- College of Engineering and Applied Sciences; Nanjing University; Nanjing 210023 P. R. China
| | - De-Shuai Lou
- Chongqing Key Laboratory of Medicinal Resources in the Three Gorges Reservoir Region; School of Biological and Chemical Engineering; Chongqing University of Education; Chongqing 400067 P. R. China
- Research Center of Brain Intellectual Promotion and Development for Children Aged 0-6 Years; Chongqing University of Education; Chongqing 400067 P. R. China
| | - Qiang Li
- Chongqing Key Laboratory of Medicinal Resources in the Three Gorges Reservoir Region; School of Biological and Chemical Engineering; Chongqing University of Education; Chongqing 400067 P. R. China
- Research Center of Brain Intellectual Promotion and Development for Children Aged 0-6 Years; Chongqing University of Education; Chongqing 400067 P. R. China
| | - Lin-Feng Hu
- Chongqing Key Laboratory of Medicinal Resources in the Three Gorges Reservoir Region; School of Biological and Chemical Engineering; Chongqing University of Education; Chongqing 400067 P. R. China
- Research Center of Brain Intellectual Promotion and Development for Children Aged 0-6 Years; Chongqing University of Education; Chongqing 400067 P. R. China
| | - Jun Tan
- Chongqing Key Laboratory of Medicinal Resources in the Three Gorges Reservoir Region; School of Biological and Chemical Engineering; Chongqing University of Education; Chongqing 400067 P. R. China
- Research Center of Brain Intellectual Promotion and Development for Children Aged 0-6 Years; Chongqing University of Education; Chongqing 400067 P. R. China
| |
Collapse
|
21
|
The Role of Thioredoxin-1 in Suppression Sepsis Through Inhibiting Mitochondrial-Induced Apoptosis in Spleen. Shock 2018; 47:753-758. [PMID: 28505020 DOI: 10.1097/shk.0000000000000789] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Sepsis is a serious public health issue and the leading cause of death in critically ill patients in intensive care units. Thioredoxin-1 (Trx-1) is a protein of regulating redox, as well as a modulator of inflammation and apoptosis. Our previous study reported that Trx-1 decreased endoplasmic reticulum-mediated inflammation involved in lung in a model of experimental sepsis. However, its effect on mitochondrial-mediated apoptosis in spleen has not been reported. We studied whether Trx-1 could prevent spleen cells apoptosis in sepsis. In the present study, we showed that the apoptosis in spleen was decreased in sepsis induced by cecal ligation and puncture (CLP) in Trx-1 overexpression transgenic (Tg) mice compared with wild-type mice. Colony forming units in the peritoneal cavity and the level of procalcitonin in plasma were significantly decreased in Trx-1 Tg mice 12 h after CLP. The expressions of c-jun-N-terminal kinase, Bax, caspase-9, and caspase-3 were increased in spleen, which were suppressed in Trx-1 Tg mice. However, the decreased Bcl-2 expression in sepsis was recovered in Trx-1 Tg mice. Our results suggest that overexpression of Trx-1 provides protection against sepsis through suppressing mitochondria-induced apoptosis pathway in spleen. This study may provide a new target for clinical intervention, as well potential strategies for treatment of sepsis.
Collapse
|
22
|
Measey TJ, Pouliot M, Wierzbicki W, Swanson C, Brown D, Authier S, Donini O. Pilot Study of Radiation-induced Gastrointestinal Injury in a Hemi-body Shielded Göttingen Minipig Model. HEALTH PHYSICS 2018; 114:43-57. [PMID: 30085969 DOI: 10.1097/hp.0000000000000751] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Development of medical countermeasures (MCMs) for gastrointestinal (GI) injury following acute radiation exposure requires well-characterized models that can assess not only survival but also secondary endpoints, including structural and functional characteristics of GI damage and recovery that ultimately contribute to long-term survival. The authors conducted a pilot study in a hemi-body shielded Göttingen minipig model of radiation-induced GI injury that enables radiation damage to the GI tract to be evaluated and reduces the potential for hemorrhage and/or damage in other more sensitive organ systems. With shielding of the head, chest, and front legs, radiation dose levels of 14 Gy were required to see significant GI-related morbidity, while dose levels of 16 Gy resulted in significant mortality by day 45 post-irradiation. Periodic scheduled necropsies showed significant reduction in and slow recovery of intestinal crypt count at 14 and 16 Gy. Intestinal proliferative activity was initially increased and then gradually decreased over the course of the study. Histological evidence of marked inflammatory infiltrates was noted in the GI tract at day 5, while collagen deposition, indicative of fibrosis, was observed as early as day 15, peaking at day 30. The radiation dose-responsive indicators of GI damage identified in this model (i.e., intestinal crypt count and proliferative activity) may serve as useful endpoints for evaluation of the efficacy of potential MCMs.
Collapse
|
23
|
Mathioudakis AG, Chatzimavridou-Grigoriadou V, Corlateanu A, Vestbo J. Procalcitonin to guide antibiotic administration in COPD exacerbations: a meta-analysis. Eur Respir Rev 2017; 26:160073. [PMID: 28143877 PMCID: PMC9488925 DOI: 10.1183/16000617.0073-2016] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 09/26/2016] [Indexed: 02/07/2023] Open
Abstract
Challenges in the differentiation of the aetiology of acute exacerbations of chronic obstructive pulmonary disease (AECOPD) have led to significant overuse of antibiotics. Serum procalcitonin, released in response to bacterial infections, but not viral infections, could possibly identify AECOPD requiring antibiotics. In this meta-analysis we assessed the clinical effectiveness of procalcitonin-based protocols to initiate or discontinue antibiotics in patients presenting with AECOPD.Based on a prospectively registered protocol, we reviewed the literature and selected randomised or quasi-randomised trials comparing procalcitonin-based protocols to initiate or discontinue antibiotics versus standard care in AECOPD. We followed Cochrane and GRADE (Grading of Recommendations, Assessment, Development and Evaluation) guidance to assess risk of bias, quality of evidence and to perform meta-analyses.We included eight trials evaluating 1062 patients with AECOPD. Procalcitonin-based protocols decreased antibiotic prescription (relative risk (RR) 0.56, 95% CI 0.43-0.73) and total antibiotic exposure (mean difference (MD) -3.83, 95% CI (-4.32--3.35)), without affecting clinical outcomes such as rate of treatment failure (RR 0.81, 0.62-1.06), length of hospitalisation (MD -0.76, -1.95-0.43), exacerbation recurrence rate (RR 0.96, 0.69-1.35) or mortality (RR 0.99, 0.58-1.69). However, the quality of the available evidence is low to moderate, because of methodological limitations and small overall study population.Procalcitonin-based protocols appear to be clinically effective; however, confirmatory trials with rigorous methodology are required.
Collapse
Affiliation(s)
- Alexander G Mathioudakis
- Division of Infection, Immunity and Respiratory Medicine, University Hospital of South Manchester, University of Manchester, Manchester, UK
| | | | - Alexandru Corlateanu
- Department of Respiratory Medicine, State University of Medicine and Pharmacy "Nicolae Testemitanu", Chisinau, Moldova
| | - Jørgen Vestbo
- Division of Infection, Immunity and Respiratory Medicine, University Hospital of South Manchester, University of Manchester, Manchester, UK
| |
Collapse
|
24
|
Neutrophil Accumulation in the Small Intestine Contributes to Local Tissue Destruction Following Combined Radiation and Burn Injury. J Burn Care Res 2016; 37:97-105. [PMID: 25501789 DOI: 10.1097/bcr.0000000000000220] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The threat of nuclear disaster makes combined radiation and burn injury (CRI) a relevant topic when discussing modern trauma, as burn injuries are likely to occur with detonation of a conventional nuclear weapon. Previous studies in a murine model have shown that there is a breakdown of the gut epithelium and subsequent bacterial translocation into mesenteric lymph nodes after CRI. This study examines the early innate immune response of the small intestine after CRI. Using a previously established murine model of 5 to 5.5 Gy total body irradiation combined with 15% TBSA burn, the injury response of the small intestine was examined at 24, 48, and 72 hours by visual assessment, myeloperoxidase, and cytokine measurement. At 24 hours, intestinal damage as measured by villus blunting, crypt debris, and decreased mitosis, was apparent in all injury groups but the derangements persisted out to 72 hours only with CRI. The prolonged intestinal damage in CRI was accompanied by a 2-fold (P < .05) elevation in myeloperoxidase activity over sham animals at 48 hours and persisted as a 3-fold (P < .05) elevation at 72 hours after injury. Corresponding levels of KC were 8-fold (P < .05) higher than sham at 48 hours with persistent elevation at 72 hours. An enhanced innate immune response, partially mediated by the influx of neutrophils into the gastrointestinal tract is contributing to the hyperinflammatory state seen after CRI. Attenuation of the local gastrointestinal inflammatory response may play a major role in managing victims after nuclear disaster.
Collapse
|
25
|
Ossetrova NI, Blakely WF, Nagy V, McGann C, Ney PH, Christensen CL, Koch AL, Gulani J, Sigal GB, Glezer EN, Hieber KP. Non-human Primate Total-body Irradiation Model with Limited and Full Medical Supportive Care Including Filgrastim for Biodosimetry and Injury Assessment. RADIATION PROTECTION DOSIMETRY 2016; 172:174-191. [PMID: 27473690 DOI: 10.1093/rpd/ncw176] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
An assessment of multiple biomarkers from radiation casualties undergoing limited- or full-supportive care including treatment with filgrastim is critical to develop rapid and effective diagnostic triage strategies. The efficacy of filgrastim with full-supportive care was compared with results with limited-supportive care by analyzing survival, necropsy, histopathology and serial blood samples for hematological, serum chemistry and protein profiles in a non-human primate (Macaca mulatta, male and female) model during 60-d post-monitoring period following sham- and total-body irradiation with 6.5 Gy 60Co gamma-rays at 0.6 Gy min-1 Filgrastim (10 μg kg-1) was administered beginning on Day 1 post-exposure and continued daily until neutrophil counts were ≥2,000 μL-1 for two consecutive days. Filgrastim and full-supportive care significantly decreased the pancytopenia duration and resulted in improved animal survival and recovery compared to animals with a limited-supportive care. These findings also identified and validated a multiparametric biomarker panel to support radiation diagnostic device development.
Collapse
Affiliation(s)
- Natalia I Ossetrova
- Scientific Research Department, Armed Forces Radiobiology Research Institute (AFRRI), Uniformed Services University (USU) , 8901 Wisconsin Avenue, Bethesda, ML 20889, USA
| | - William F Blakely
- Scientific Research Department, Armed Forces Radiobiology Research Institute (AFRRI), Uniformed Services University (USU) , 8901 Wisconsin Avenue, Bethesda, ML 20889, USA
| | - Vitaly Nagy
- Scientific Research Department, Armed Forces Radiobiology Research Institute (AFRRI), Uniformed Services University (USU) , 8901 Wisconsin Avenue, Bethesda, ML 20889, USA
| | - Camille McGann
- Scientific Research Department, Armed Forces Radiobiology Research Institute (AFRRI), Uniformed Services University (USU) , 8901 Wisconsin Avenue, Bethesda, ML 20889, USA
| | - Patrick H Ney
- Scientific Research Department, Armed Forces Radiobiology Research Institute (AFRRI), Uniformed Services University (USU) , 8901 Wisconsin Avenue, Bethesda, ML 20889, USA
| | - Christine L Christensen
- Scientific Research Department, Armed Forces Radiobiology Research Institute (AFRRI), Uniformed Services University (USU) , 8901 Wisconsin Avenue, Bethesda, ML 20889, USA
- Tri-Service Research Laboratory (TRSL), 4141 Petroleum Road, JBSA-Fort Sam Houston, TX 78234, USA
| | - Amory L Koch
- Scientific Research Department, Armed Forces Radiobiology Research Institute (AFRRI), Uniformed Services University (USU) , 8901 Wisconsin Avenue, Bethesda, ML 20889, USA
- Tripler Army Medical Center, Honolulu, HI 96859, USA
| | - Jatinder Gulani
- Scientific Research Department, Armed Forces Radiobiology Research Institute (AFRRI), Uniformed Services University (USU) , 8901 Wisconsin Avenue, Bethesda, ML 20889, USA
| | - George B Sigal
- Meso Scale Diagnostics, LLC. (MSD), 1601 Research Boulevard, Rockville, MD 20850, USA
| | - Eli N Glezer
- Meso Scale Diagnostics, LLC. (MSD), 1601 Research Boulevard, Rockville, MD 20850, USA
| | - Kevin P Hieber
- Scientific Research Department, Armed Forces Radiobiology Research Institute (AFRRI), Uniformed Services University (USU) , 8901 Wisconsin Avenue, Bethesda, ML 20889, USA
| |
Collapse
|
26
|
Garg S, Zheng J, Wang J, Authier S, Pouliot M, Hauer-Jensen M. Segmental Differences in Radiation-Induced Alterations of Tight Junction-Related Proteins in Non-Human Primate Jejunum, Ileum and Colon. Radiat Res 2015; 185:50-9. [PMID: 26720804 DOI: 10.1667/rr14157.1] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Dysfunction of the intestinal epithelial barrier and leakage of luminal antigens and bacteria across the barrier have been linked to various human diseases. Intestinal permeability is regulated by intercellular structures, termed "tight junction" (Tj), which are disrupted after total-body irradiation (TBI). In this study, we investigated radiation-induced alterations in Tj-related proteins in the jejunum, ileum and colon of a non-human primate (NHP) model. NHPs were total-body irradiated with 6.7 and 7.4 Gy and intestines were procured at day 4, 7 and 12. Radiation exposure was found to induce significant increases in claudin-10 mRNA early (day 4) in all three gut segments and claudin-4 mRNA levels were repressed through day 12. TNF-alpha was highly induced in the jejunum and colon at early time points, but little induction was found in the ileum. Claudin-1 was induced only in the colon on day 4 postirradiation. Unlike the colon and jejunum, the ileum levels of claudin-7 were significantly downregulated through day 12 postirradiation. Western blot analysis revealed increased levels of claudin-2 on day 4 and of JAM-1 on day 7 postirradiation in all three gut segments. E-cadherin was downregulated on day 4 postirradiation in all segments, but remained reduced in the jejunum only until day 12. Taken together, these data suggest that exposure to radiation causes segment-specific alterations in the expression of Tj-related proteins. Interruption of Tjs may be a key factor contributing to injury to the intestinal mucosal barrier and increased intestinal permeability.
Collapse
Affiliation(s)
- Sarita Garg
- a Department of Pharmaceutical Sciences, Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Junying Zheng
- a Department of Pharmaceutical Sciences, Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Junru Wang
- a Department of Pharmaceutical Sciences, Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | | | | | - Martin Hauer-Jensen
- a Department of Pharmaceutical Sciences, Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, Arkansas;,c Surgical Service, Central Arkansas Veterans Healthcare System, Little Rock, Arkansas
| |
Collapse
|
27
|
Zheng J, Wang J, Pouliot M, Authier S, Zhou D, Loose DS, Hauer-Jensen M. Gene expression profiling in non-human primate jejunum, ileum and colon after total-body irradiation: a comparative study of segment-specific molecular and cellular responses. BMC Genomics 2015; 16:984. [PMID: 26589571 PMCID: PMC4654820 DOI: 10.1186/s12864-015-2168-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Accepted: 10/29/2015] [Indexed: 12/26/2022] Open
Abstract
Background Although extensive studies have investigated radiation-induced injuries in particular gastrointestinal (GI) segments, a systematic comparison among the different segments on the basis of mode, magnitude and mechanism has not been reported. Here, a comparative study of segment-specific molecular and cellular responses was performed on jejunum, ileum and colon obtained at three time points (4, 7 and 12 days after irradiation) from non-human primate (Rhesus macaque) models exposed to 6.7 Gy or 7.4 Gy total body irradiation (TBI). Results Pathway analysis on the gene expression profiles identified radiation-induced time-, dose- and segment-dependent activation of tumor necrosis factor α (TNFα) cascade, tight junction, apoptosis, cell cycle control/DNA damage repair and coagulation system signaling. Activation of these signaling pathways suggests that colon sustained the severest mucosal barrier disruption and inflammation, and jejunum the greatest DNA damage, apoptosis and endothelial dysfunction. These more pronounced alterations correlate with the high incidence of macroscopic pathologies that are observed in the colon after TBI. Compared to colon and jejunum, ileum was resistant to radiation injury. In addition to the identification a marked increase of TNFα cascade, this study also identified radiation induced strikingly up-regulated tight junction gene CLDN2 (196-fold after 7.4-Gy TBI), matrix degradation genes such as MMP7 (increased 11- and 41-fold after 6.7-Gy and 7.4-Gy TBI), and anoikis mediated gene EDA2R that mediate mucosal shedding and barrier disruption. Conclusions This is the first systematic comparative study of the molecular and cellular responses to radiation injury in jejunum, ileum and colon. The strongest activation of TNFα cascades and the striking up-regulation of its down-stream matrix-dissociated genes suggest that TNFα modulation could be a target for mitigating radiation-induced mucosal barrier disruption. Electronic supplementary material The online version of this article (doi:10.1186/s12864-015-2168-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Junying Zheng
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, Arkansas, 72205, USA.
| | - Junru Wang
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, Arkansas, 72205, USA.
| | | | - Simon Authier
- CiToxLAB North America, Laval, Quebec, Canada, H7V 4B3.
| | - Daohong Zhou
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, Arkansas, 72205, USA.
| | - David S Loose
- Integrative Biology and Pharmacology, University of Texas Medical School at Houston, Houston, TX, 77030, USA.
| | - Martin Hauer-Jensen
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, Arkansas, 72205, USA. .,Surgical Service, Central Arkansas Veterans Healthcare System, Little Rock, Arkansas, 72205, USA.
| |
Collapse
|
28
|
de Oliveira THC, Amorin AT, Rezende IS, Santos Barbosa M, Martins HB, Brito AKP, Andrade EF, Gonçalves GKN, Campos GB, Silva RAA, Timenetsky J, Marques LM. Sepsis induced by Staphylococcus aureus: participation of biomarkers in a murine model. Med Sci Monit 2015; 21:345-55. [PMID: 25630550 PMCID: PMC4321564 DOI: 10.12659/msm.892528] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND This study aimed to evaluate the role of biomarkers in the pathophysiological process induced by a Staphylococcus aureus strain obtained in a hospital environment. For this, we intraperitoneally inoculated groups of male BALB/c mice with S. aureus, using a clinical isolate (CI) of S. aureus. MATERIAL/METHODS Mice were divided into groups according to time of euthanasia (24, 48, 72, 96, 120, 144, and 168 hours of infection). After being euthanized, blood samples were collected for quantification of microorganisms and leukocytes, as well as measurement of biomarkers of tumor necrosis factor alpha (TNF-α), interleukin 6 (IL-6), C-reactive protein (CRP), and Procalcitonin (PCT) by ELISA. Heart, kidneys, and lungs were removed for histopathological analysis, assessment of biomarkers of tissue expression by RT-PCR (polymerase chain reaction with reverse transcriptase), and quantification of microorganisms by real-time quantitative PCR (real-time PCR). RESULTS The animals infected at between 120 hours and 168 hours had the highest blood levels of S. aureus. We observed that infection promoted increases in the levels of circulating neutrophils and monocytes. However, there was a reduction of circulating neutrophils and monocytes after 96 hours of infection. The infected mice also had increased levels of blood lymphocytes. In this model of infection with S. aureus, IL-6, CRP, and PCT demonstrated greater fidelity as markers of infection, since serum levels were elevated and lowered along with the number of circulating neutrophils and monocytes after resolution of the infection. The lungs showed hyperemia, with enlargement of the alveolar septa. On the other hand, infection with S. aureus did not promote visible change in histological tissue in the heart and kidneys. CONCLUSIONS In this model of infection with S. aureus, IL-6, CRP, and PCT demonstrated greater fidelity as markers of infection, since serum levels were elevated and lowered along with the number of circulating neutrophils and monocytes after resolution of the infection. We believe our results may provide a better understanding of the pathophysiology, as well as aid in the search for a more reliable method of diagnosis.
Collapse
Affiliation(s)
| | - Aline Teixeira Amorin
- Multidisciplinary Institute of Health, Federal University of Bahia, Vitória da Conquista, Brazil
| | - Izadora Souza Rezende
- Multidisciplinary Institute of Health, Federal University of Bahia, Vitória da Conquista, Brazil
| | - Maysa Santos Barbosa
- Multidisciplinary Institute of Health, Federal University of Bahia, Vitória da Conquista, Brazil
| | - Hellen Braga Martins
- Multidisciplinary Institute of Health, Federal University of Bahia, Vitória da Conquista, Brazil
| | | | - Ewerton Ferraz Andrade
- Multidisciplinary Institute of Health, Federal University of Bahia, Vitória da Conquista, Brazil
| | | | - Guilherme Barreto Campos
- Department of Microbiologia, Biomedical Science Institute, São Paulo University, São Paulo, Brazil
| | | | - Jorge Timenetsky
- Department of Microbiologia, Biomedical Science Institute, São Paulo University, São Paulo, Brazil
| | - Lucas Miranda Marques
- Multidisciplinary Institute of Health, Federal University of Bahia, Vitória da Conquista, Brazil
| |
Collapse
|