Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 101
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Article in English | MEDLINE | ID: mdl-38234297

ABSTRACT

Background. Silica nanoparticles found in sugarcane ash have been postulated to be a toxicant contributing to chronic kidney disease of unknown etiology (CKDu). However, while the administration of manufactured silica nanoparticles is known to cause chronic tubulointerstitial disease in rats, the effect of administering sugarcane ash on kidney pathology remains unknown. Here we investigate whether sugarcane ash can induce CKD in rats. Methods. Sugarcane ash was administered for 13 weeks into the nares of rats (5 mg/day for 5d/week), and blood, urine and kidney tissues were collected at 13 weeks (at the end of ash administration) and in a separate group of rats at 24 weeks (11 weeks after stopping ash administration). Kidney histology was evaluated, and inflammation and fibrosis (collagen deposition) measured. Results. Sugarcane ash exposure led to the accumulation of silica in the kidneys, lungs, liver and spleen of rats. Mild proteinuria developed although renal function was largely maintained. However, biopsies showed focal glomeruli with segmental glomerulosclerosis, and tubulointerstitial inflammation and fibrosis that tended to worsen even after the ash administration had been stopped. Staining for the lysosomal marker, LAMP-1, showed decreased staining in ash administered rats consistent with lysosomal activation. Conclusion. Sugarcane ash containing silica nanoparticles can cause CKD in rats.

2.
Inhal Toxicol ; : 1-16, 2024 Feb 13.
Article in English | MEDLINE | ID: mdl-38349733

ABSTRACT

Sugarcane is the most widely cultivated crop in the world, with equatorial developing nations performing most of this agriculture. Burning sugarcane is a common practice to facilitate harvest, producing extremely high volumes of respirable particulate matter in the process. These emissions are known to have deleterious effects on agricultural workers and nearby communities, but the extent of this exposure and potential toxicity remain poorly characterized. As the epidemicof chronic kidney disease of an unknown etiology (CKDu) and its associated mortality continue to increase along with respiratory distress, there is an urgent need to investigate the causes, determine viable interventions to mitigate disease andimprove outcomes for groups experiencing disproportionate impact. The goal of this review is to establish the state of available literature, summarize what is known in terms of human health risk, and provide recommendations for what areas should be prioritized in research.

3.
Nephrol Dial Transplant ; 38(1): 41-48, 2023 Jan 23.
Article in English | MEDLINE | ID: mdl-34473287

ABSTRACT

Climate change should be of special concern for the nephrologist, as the kidney has a critical role in protecting the host from dehydration, but it is also a favorite target of heat stress and dehydration. Here we discuss how rising temperatures and extreme heat events may affect the kidney. The most severe presentation of heat stress is heat stroke, which can result in severe electrolyte disturbance and both acute and chronic kidney disease (CKD). However, lesser levels of heat stress also have multiple effects, including exacerbating kidney disease and precipitating cardiovascular events in subjects with established kidney disease. Heat stress can also increase the risk for kidney stones, cause multiple electrolyte abnormalities and induce both acute and chronic kidney disease. Recently there have been multiple epidemics of CKD of uncertain etiology in various regions of the world, including Mesoamerica, Sri Lanka, India and Thailand. There is increasing evidence that climate change and heat stress may play a contributory role in these conditions, although other causes, including toxins, could also be involved. As climate change worsens, the nephrologist should prepare for an increase in diseases associated with heat stress and dehydration.


Subject(s)
Heat Stress Disorders , Nephrology , Renal Insufficiency, Chronic , Humans , Climate Change , Dehydration/complications , Renal Insufficiency, Chronic/complications , Kidney , Heat Stress Disorders/complications
4.
J Immunol ; 207(11): 2637-2648, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34732470

ABSTRACT

Mast cells are important effector cells in the immune system and undergo activation (i.e., degranulation) by two major mechanisms: IgE-mediated and non-IgE-mediated mechanisms. Although IgE-mediated degranulation is well researched, the cellular mechanisms of non-IgE-mediated mast cell activation are poorly understood despite the potential to induce similar pathophysiological effects. To better understand non-IgE mast cell degranulation, we characterized and compared cellular metabolic shifts across several mechanisms of degranulation (allergen-induced [IgE-mediated], 20 nm of silver nanoparticle-mediated [non-IgE], and compound 48/80-mediated [non-IgE]) in murine bone marrow-derived mast cells. All treatments differentially impacted mitochondrial activity and glucose uptake, suggesting diverging metabolic pathways between IgE- and non-IgE-mediated degranulation. Non-IgE treatments depleted mast cells' glycolytic reserve, and compound 48/80 further inhibited the ability to maximize mitochondrial respiration. This cellular reprogramming may be indicative of a stress response with non-IgE treatments. Neither of these outcomes occurred with IgE-mediated degranulation, hinting at a separate programmed response. Fuel flexibility between the three primary mitochondrial nutrient sources was also eliminated in activated cells and this was most significant in non-IgE-mediated degranulation. Lastly, metabolomics analysis of bone marrow-derived mast cells following degranulation was used to compare general metabolite profiles related to energetic pathways. IgE-mediated degranulation upregulated metabolite concentrations for the TCA cycle and glycolysis compared with other treatments. In conclusion, mast cell metabolism varies significantly between IgE- and non-IgE-mediated degranulation suggesting novel cell regulatory mechanisms are potentially driving unexplored pathways of mast cell degranulation.


Subject(s)
Immunoglobulin E/metabolism , Mast Cells/metabolism , Animals , Cells, Cultured , Immunoglobulin E/immunology , Mast Cells/immunology , Mice , Mice, Inbred C57BL
5.
Environ Res ; 239(Pt 1): 117311, 2023 Dec 15.
Article in English | MEDLINE | ID: mdl-37805178

ABSTRACT

EXPOSURE TO POLY: and perfluoroalkyl substances (PFAS) in early life may increase the risk of childhood asthma, but evidence has been inconsistent. We estimated associations between maternal serum concentrations of PFAS during pregnancy and clinician-diagnosed asthma incidence in offspring through age eight. We included 597 mother-child pairs with PFAS quantified in mid-pregnancy serum and childhood medical records reviewed for asthma diagnoses. We used separate Cox proportional hazards models to assess the relationship between log-transformed concentrations of five PFAS and the incidence of asthma. We estimated associations between the PFAS mixture and clinician-diagnosed asthma incidence using quantile-based g-computation. PFAS concentrations were similar to those among females in the US general population. Seventeen percent of children (N = 104) were diagnosed with asthma during follow-up. Median (interquartile range) duration of follow-up was 4.7 (4.0, 6.2) years, and median age at asthma diagnosis was 1.7 (0.9, 2.8) years. All adjusted hazard ratios (HRs) were elevated, but all 95% confidence intervals (CI) included the null. The HR (95% CI) of asthma for a one-quartile increase in the PFAS mixture was 1.17 (0.86, 1.61). In this cohort of children followed to eight years of age, prenatal PFAS concentrations were not significantly associated with incidence of clinician-diagnosed asthma.


Subject(s)
Asthma , Fluorocarbons , Prenatal Exposure Delayed Effects , Female , Pregnancy , Humans , Child, Preschool , Incidence , Prenatal Exposure Delayed Effects/chemically induced , Prenatal Exposure Delayed Effects/epidemiology , Asthma/chemically induced , Asthma/epidemiology , Family , Fluorocarbons/toxicity
6.
Am J Physiol Renal Physiol ; 323(1): F48-F58, 2022 07 01.
Article in English | MEDLINE | ID: mdl-35635324

ABSTRACT

Silica nanoparticles (SiNPs) released during the burning of sugarcane have been postulated to have a role in chronic kidney disease of unknown etiology. We tested the hypothesis that pristine SiNPs of the size present in sugarcane might cause chronic kidney injury when administered through the lung in rats. We administered 200- or 300-nm amorphous SiNPs twice weekly (4 mg/dose), or vehicle by oropharyngeal aspiration for 13 wk to rats followed by euthanasia after an additional 13 wk (26 wk total). Tissues were evaluated for the presence of SiNPs and evidence of histological injury. Both sizes of SiNPs caused kidney damage, with early tubular injury and inflammation (at week 13) that continued to inflammation and chronic fibrosis at week 26 despite discontinuation of the SiNP administration. Both sizes of SiNPs caused local inflammation in the lung and kidney and were detected in the serum and urine at week 13, and the 200-nm particles were also localized to the kidney with no evidence of retention of the 300-nm particles. At week 26, there was some clearance of the 200-nm silica from the kidneys, and urinary levels of SiNPs were reduced but still significant in both 200- and 300 nm-exposed rats. In conclusion, inhaled SiNPs cause chronic kidney injury that progresses despite stopping the SiNP administration. These findings support the hypothesis that human exposure to amorphous silica nanoparticles found in burned sugarcane fields could have a participatory role in chronic kidney disease of unknown etiology.NEW & NOTEWORTHY Inhalation of silica nanoparticles (SiNPs) released during the burning of sugarcane has been postulated to have a role in chronic kidney disease of unknown etiology (CKDu). We administered 200- and 300-nm amorphous SiNPs to rats by aspiration and observed kidney damage with tubular injury and inflammation that persisted even after stopping the SiNP exposure. These findings support the hypothesis that human exposure to SiNPs found in sugarcane ash could have a participatory role CKDu.


Subject(s)
Nanoparticles , Renal Insufficiency, Chronic , Animals , Inflammation/pathology , Lung/pathology , Nanoparticles/toxicity , Rats , Renal Insufficiency, Chronic/chemically induced , Renal Insufficiency, Chronic/pathology , Silicon Dioxide/toxicity
7.
Inhal Toxicol ; 34(13-14): 412-432, 2022.
Article in English | MEDLINE | ID: mdl-36394251

ABSTRACT

Over 40% of veterans from the Persian Gulf War (GW) (1990-1991) suffer from Gulf War Illness (GWI). Thirty years since the GW, the exposure and mechanism contributing to GWI remain unclear. One possible exposure that has been attributed to GWI are chemical warfare agents (CWAs). While there are treatments for isolated symptoms of GWI, the number of respiratory and cognitive/neurological issues continues to rise with minimum treatment options. This issue does not only affect veterans of the GW, importantly these chronic multisymptom illnesses (CMIs) are also growing amongst veterans who have served in the Afghanistan-Iraq war. What both wars have in common are their regions and inhaled exposures. In this review, we will describe the CWA exposures, such as sarin, cyclosarin, and mustard gas in both wars and discuss the various respiratory and neurocognitive issues experienced by veterans. We will bridge the respiratory and neurological symptoms experienced to the various potential mechanisms described for each CWA provided with the most up-to-date models and hypotheses.


Subject(s)
Chemical Warfare Agents , Persian Gulf Syndrome , Veterans , Humans , Chemical Warfare Agents/toxicity , Persian Gulf Syndrome/chemically induced , Gulf War , Sarin
9.
Toxicol Appl Pharmacol ; 382: 114746, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31494149

ABSTRACT

Mast cells are a key effector cell in type I allergic reactions. It has been shown that environmental exposures such as diesel exhaust and heavy metals exacerbate mast cell degranulation and activation. Today, the use of engineered nanomaterials (ENMs) is rapidly expanding and silver nanoparticles (AgNP) are one of the mostly widely utilized ENMs, primarily for their antimicrobial properties, and are being incorporated into many consumer and biomedical products. We assessed whether pre-exposure of bone marrow-derived mast cells (BMMCs) to 20 nm AgNPs enhanced degranulation and activation to an allergen (dinitrophenol-conjugated human serum albumin) by measuring ß-hexosaminidase release, LTB4 and IL-6 production. In addition, we assessed reactive oxygen species (ROS) generation, cell oxidative stress and toxicity as well as total and individual protein tyrosine phosphorylation (p-Tyr). We found that pre-exposure of BMMCs to AgNPs results in exacerbated allergen-mediated mast cell degranulation, LTB4 production and IL-6 release. Exposure of BMMCs to AgNPs exacerbated allergen-induced ROS generation, however, this was not associated with oxidative stress nor cell death. Finally, pre-exposure to AgNPs enhanced allergen-mediated global p-Tyr as well as individual proteins including Syk, PLCγ and LAT. Our findings indicate that pre-exposure to AgNPs exacerbates mast cell allergen-mediated phosphorylation of FcεR1-linked tyrosine kinases and ROS generation resulting in amplified early and late-phase responses. These findings suggest that exposure to AgNPs has the potential to prime mast cells to allergic immune responses, which could be of particular concern to atopic populations as the use of AgNPs in consumer and biomedical products rapidly increases.


Subject(s)
Mast Cells/drug effects , Mast Cells/metabolism , Metal Nanoparticles/toxicity , Receptors, IgE/metabolism , Silver/toxicity , Animals , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Mast Cells/immunology , Mice , Mice, Inbred C57BL , Receptors, IgE/agonists , Receptors, IgE/immunology
10.
Ecotoxicol Environ Saf ; 170: 77-86, 2019 Apr 15.
Article in English | MEDLINE | ID: mdl-30529623

ABSTRACT

Prior research has demonstrated cells exposed to silver nanoparticles (AgNPs) undergo endoplasmic reticulum (ER) stress leading to cellular apoptosis and toxicity, however, the fundamental mechanism underlying AgNP-induced ER stress is unknown. We hypothesize the biophysical interactions between AgNPs and adsorbed proteins lead to misfolded proteins to elicit an ER stress response. Our investigation examined rat aortic endothelial cells (RAEC) exposed to 20 or 100 nm AgNPs with or without a biocorona (BC) consisting of bovine serum albumin (BSA), high density lipoprotein (HDL) or fetal bovine serum (FBS) to form a complex BC. The presence of a BC consisting of BSA or FBS proteins significantly reduced uptake of 20 nm and 100 nm AgNPs in RAEC. Western blot analysis indicated robust activation of the IREα and PERK pathways in RAEC exposed to 20 nm despite the reduction in uptake by the presence of a BC. This was not observed for the 100 nm AgNPs. Hyperspectral darkfield microscopy qualitatively confirmed that the preformed BC was maintained following uptake by RAEC. Transmission electron microscopy demonstrated a size dependent effect on the sub-cellular localization of AgNPs. Overall, these results suggest that AgNP size, surface area and BC formation governs the induction of ER stress and alterations in intracellular trafficking.


Subject(s)
Endoplasmic Reticulum Stress/drug effects , Metal Nanoparticles/toxicity , Protein Corona , Silver/toxicity , Adsorption , Animals , Apoptosis/drug effects , Cell Survival/drug effects , Cells, Cultured , Circular Dichroism , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Lipoproteins, HDL/chemistry , Microscopy, Electron, Transmission , Oxidative Stress/drug effects , Rats , Serum Albumin, Bovine/chemistry
11.
J Hepatol ; 2017 Sep 21.
Article in English | MEDLINE | ID: mdl-28870670

ABSTRACT

BACKGROUND & AIM: Alcohol consumption increases intestinal permeability and causes damage to hepatocytes, leading to the release of pathogen- and damage-associated molecular pattern molecules (PAMPs and DAMPs), stimulating hepatic macrophages and activating NF-κB. The resultant inflammation exacerbates alcoholic liver disease (ALD). However, much less is known about the mechanisms attenuating inflammation and preventing disease progression in most heavy drinkers. Interleukin (IL)-33 is a DAMP (alarmin) released from dead cells that acts through its receptor, IL-1 receptor like 1 (ST2). ST2 signaling has been reported to either stimulate or inhibit NF-κB activation. The role of IL-33/ST2 in ALD has not been studied. METHODS: Serum levels of IL-33 and its decoy receptor, soluble ST2 (sST2) were measured in ALD patients. Alcohol-induced liver injury, inflammation and hepatic macrophage activation were compared between wild-type, IL-33-/- and ST2-/- mice in several models. RESULTS: Elevation of serum IL-33 and sST2 were only observed in patients with severe decompensated ALD. Consistently, in mice with mild ALD without significant cell death and IL-33 release, IL-33 deletion did not affect alcohol-induced liver damage. However, ST2-deletion exacerbated ALD, through enhancing NF-κB activation in liver macrophages. In contrast, when extracellular IL-33 was markedly elevated, liver injury and inflammation were attenuated in both IL-33-/- and ST2-/- mice compared to wild-type mice. CONCLUSION: Our data revealed a dichotomous role of IL-33/ST2 signaling during ALD development. At early and mild stages, ST2 restrains the inflammatory activation of hepatic macrophages, through inhibiting NF-κB, and plays a protective function in an IL-33-independent fashion. During severe liver injury, significant cell death and marked IL-33 release occur, which triggers IL-33/ST2 signaling and exacerbates tissue damage. LAY SUMMARY: In mild ALD, ST2 negatively regulates the inflammatory activation of hepatic macrophages, thereby protecting against alcohol-induced liver damage, whereas in the case of severe liver injury, the release of extracellular IL-33 may exacerbate tissue inflammation by triggering the canonical IL-33/ST2L signaling in hepatic macrophages.

12.
Am J Physiol Lung Cell Mol Physiol ; 310(2): L142-54, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26589480

ABSTRACT

Pulmonary instillation of multiwalled carbon nanotubes (MWCNT) has the potential to promote cardiovascular derangements, but the mechanisms responsible are currently unclear. We hypothesized that exposure to MWCNT would result in increased epithelial barrier permeability by 24 h postexposure and initiate a signaling process involving IL-6/gp130 transsignaling in peripheral vascular tissue. To test this hypothesis we assessed the impact of 1 and 10 µg/cm(2) MWCNT on transepithelial electrical resistance (TEER) and expression of barrier proteins and cell activation in vitro using normal human bronchial epithelial primary cells. Parallel studies using male Sprague-Dawley rats instilled with 100 µg MWCNT measured bronchoalveolar lavage (BAL) differential cell counts, BAL fluid total protein, and lung water-to-tissue weight ratios 24 h postexposure and quantified serum concentrations of IL-6, soluble IL-6r, and soluble gp130. Aortic sections were examined immunohistochemically for gp130 expression, and gp130 mRNA/protein expression was evaluated in rat lung, heart, and aortic tissue homogenates. Our in vitro findings indicate that 10 µg/cm(2) MWCNT decreased the development of TEER and zonula occludens-1 expression relative to the vehicle. In rats MWCNT instillation increased BAL protein, lung water, and induced pulmonary eosinophilia. Serum concentrations of soluble gp130 decreased, aortic endothelial expression of gp130 increased, and expression of gp130 in the lung was downregulated in the MWCNT-exposed group. We propose that pulmonary exposure to MWCNT can manifest as a reduced epithelial barrier and activator of vascular gp130-associated transsignaling that may promote susceptibility to cardiovascular derangements.


Subject(s)
Coronary Vessels/metabolism , Cytokine Receptor gp130/metabolism , Interleukin-6/metabolism , Lung/metabolism , Nanotubes, Carbon , Animals , Bronchoalveolar Lavage Fluid/cytology , Cells, Cultured , Humans , Male , Permeability , Pulmonary Artery/metabolism , Rats, Sprague-Dawley , Signal Transduction
13.
Immunology ; 146(3): 470-85, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26288256

ABSTRACT

Human mast cells (huMCs) are involved in both innate and adaptive immune responses where they release mediators including amines, reactive oxygen species (ROS), eicosanoids and cytokines. We have reported that interferon-γ (IFN-γ) enhances FcγR-dependent ROS production. The aim of this study was to extend these observations by investigating the effect of IFN-γ on the biological responses of huMCs to Staphylococcus aureus. We found that exposure of huMCs to S. aureus generated intracellular and extracellular ROS, which were enhanced in the presence of IFN-γ. IFN-γ also promoted bacteria killing, ß-hexosaminidase release and eicosanoid production. Interferon-γ similarly increased expression of mRNAs encoding CCL1 to CCL4, granulocyte-macrophage colony-stimulating factor (GM-CSF), tumour necrosis factor-α and CXCL8 in S. aureus-stimulated huMCs. The ability of IFN-γ to increase CXCL8 and GM-CSF protein levels was confirmed by ELISA. Fibronectin or a ß1 integrin blocking antibody completely abrogated IFN-γ-dependent S. aureus binding and reduced S. aureus-dependent CXCL8 secretion. These data demonstrate that IFN-γ primes huMCs for enhanced anti-bacterial and pro-inflammatory responses to S. aureus, partially mediated by ß1 integrin.


Subject(s)
Interferon-gamma/immunology , Mast Cells/immunology , Mast Cells/microbiology , Staphylococcus aureus/immunology , Adaptive Immunity , Animals , Cells, Cultured , Chemokines, CC/genetics , Chemokines, CC/metabolism , Cytokines/genetics , Cytokines/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Immunity, Innate , Inflammation Mediators/metabolism , Integrin beta1/metabolism , Interferon-gamma/pharmacology , Interleukin-8/genetics , Interleukin-8/metabolism , Mast Cells/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reactive Oxygen Species/metabolism , Staphylococcus aureus/pathogenicity , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
14.
J Immunol ; 190(2): 531-8, 2013 Jan 15.
Article in English | MEDLINE | ID: mdl-23248261

ABSTRACT

IL-33 is elevated in afflicted tissues of patients with mast cell (MC)-dependent chronic allergic diseases. Based on its acute effects on mouse MCs, IL-33 is thought to play a role in the pathogenesis of allergic disease through MC activation. However, the manifestations of prolonged IL-33 exposure on human MC function, which best reflect the conditions associated with chronic allergic disease, are unknown. In this study, we found that long-term exposure of human and mouse MCs to IL-33 results in a substantial reduction of MC activation in response to Ag. This reduction required >72 h exposure to IL-33 for onset and 1-2 wk for reversion following IL-33 removal. This hyporesponsive phenotype was determined to be a consequence of MyD88-dependent attenuation of signaling processes necessary for MC activation, including Ag-mediated calcium mobilization and cytoskeletal reorganization, potentially as a consequence of downregulation of the expression of phospholipase Cγ(1) and Hck. These findings suggest that IL-33 may play a protective, rather than a causative, role in MC activation under chronic conditions and, furthermore, reveal regulated plasticity in the MC activation phenotype. The ability to downregulate MC activation in this manner may provide alternative approaches for treatment of MC-driven disease.


Subject(s)
Immunosuppression Therapy , Interleukins/immunology , Mast Cells/immunology , Mast Cells/metabolism , Phenotype , Actins/metabolism , Animals , Bone Marrow Cells/drug effects , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Humans , Interleukin-1 Receptor-Like 1 Protein , Interleukin-33 , Interleukins/pharmacology , Mast Cells/drug effects , Mice , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/immunology , Phospholipase C gamma/genetics , Phospholipase C gamma/immunology , Phospholipase C gamma/metabolism , Protein Multimerization/drug effects , Proto-Oncogene Proteins c-hck/genetics , Proto-Oncogene Proteins c-hck/immunology , Receptors, Interleukin/genetics , Receptors, Interleukin/immunology
15.
Inhal Toxicol ; 26(4): 240-9, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24502429

ABSTRACT

OBJECTIVE: Instillation of multi-walled carbon nanotubes (MWCNTs) in C57BL/6 mice results in decrements of pulmonary function specifically characterized by increases in airway resistance. In this study, we examined possible mechanisms responsible for these alterations following MWCNT exposure, including the roles of IL-33 and chronic inflammation. MATERIALS AND METHODS: To elucidate the role of IL-33, we assessed lung histology and pulmonary function in C57BL/6 and IL-33(-/-) mice 30 days following MWCNT instillation. In addition, the impact of MWCNT instillation on airway hyperresponsiveness (AHR) was assessed by methacholine challenges of C57BL/6 and IL-33(-/-) mice. To further understand the mechanisms by which MWCNTs may increase airway constriction, C57BL/6 mice were treated with aerosolized albuterol or injected with multiple doses of methylprednisolone via intra-peritoneal injections prior to the assessment of MWCNT-induced changes in pulmonary function. RESULTS: Total cell count, macrophages, and neutrophils were increased in the lavage fluid of C57BL/6 mice, but not in IL-33(-/-) mice, following MWCNT exposure. C57BL/6 mice displayed increased inflammation and fibrosis located proximal to the airways which was absent in IL-33(-/-) mice. Aerosolized methacholine increased parameters of airway resistance (R and Rn) in a dose-dependent manner in all groups, with MWCNT-instilled C57BL/6 mice responding more robustly compared to the controls, while no differences were found in IL-33(-/-) mice due to MWCNT exposure. Treatment with methylprednisolone reduced both the MWCNT-induced histopathological changes and increases in R and Rn in C57BL/6 mice. CONCLUSION: These findings suggest that IL-33 and chronic inflammation in general are critical in the pulmonary toxicity induced by MWCNT resulting in modified pulmonary function.


Subject(s)
Airway Resistance/physiology , Interleukins/physiology , Lung/drug effects , Nanotubes, Carbon/toxicity , Adrenergic beta-2 Receptor Agonists/pharmacology , Albuterol/pharmacology , Animals , Bronchial Provocation Tests , Bronchoalveolar Lavage Fluid/cytology , Cell Count , Glucocorticoids/pharmacology , Interleukin-33 , Interleukins/genetics , Lung/pathology , Lung/physiopathology , Male , Methylprednisolone/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout
16.
Kidney Int Rep ; 9(5): 1458-1472, 2024 May.
Article in English | MEDLINE | ID: mdl-38707825

ABSTRACT

Introduction: Sugarcane workers are exposed to potentially hazardous agrochemicals, including pesticides, heavy metals, and silica. Such occupational exposures present health risks and have been implicated in a high rate of kidney disease seen in these workers. Methods: To investigate potential biomarkers and mechanisms that could explain chronic kidney disease (CKD) among this worker population, paired urine samples were collected from sugarcane cutters at the beginning and end of a harvest season in Guatemala. Workers were then separated into 2 groups, namely those with or without kidney function decline (KFD) across the harvest season. Urine samples from these 2 groups underwent elemental analysis and untargeted metabolomics. Results: Urine profiles demonstrated increases in silicon, certain pesticides, and phosphorus levels in all workers, whereas heavy metals remained low. The KFD group had a reduction in estimated glomerular filtration rate (eGFR) across the harvest season; however, kidney injury marker 1 did not significantly change. Cross-harvest metabolomic analysis found trends of fatty acid accumulation, perturbed amino acid metabolism, presence of pesticides, and other known signs of impaired kidney function. Conclusion: Silica and certain pesticides were significantly elevated in the urine of sugarcane workers with or without KFD. Future work should determine whether long-term occupational exposure to silica and pesticides across multiple seasons contributes to CKD in these workers. Overall, these results confirmed that multiple exposures are occurring in sugarcane workers and may provide insight into early warning signs of kidney injury and may help explain the increased incidence of CKD among agricultural workers.

17.
ACS Chem Neurosci ; 15(7): 1533-1547, 2024 04 03.
Article in English | MEDLINE | ID: mdl-38507813

ABSTRACT

Neuroinflammation plays a crucial role in the development of neurodegenerative protein misfolding disorders. This category of progressive diseases includes, but is not limited to, Alzheimer's disease, Parkinson's disease, and prion diseases. Shared pathogenesis involves the accumulation of misfolded proteins, chronic neuroinflammation, and synaptic dysfunction, ultimately leading to irreversible neuronal loss, measurable cognitive deficits, and death. Presently, there are few to no effective treatments to halt the advancement of neurodegenerative diseases. We hypothesized that directly targeting neuroinflammation by downregulating the transcription factor, NF-κB, and the inflammasome protein, NLRP3, would be neuroprotective. To achieve this, we used a cocktail of RNA targeting therapeutics (SB_NI_112) shown to be brain-penetrant, nontoxic, and effective inhibitors of both NF-κB and NLRP3. We utilized a mouse-adapted prion strain as a model for neurodegenerative diseases to assess the aggregation of misfolded proteins, glial inflammation, neuronal loss, cognitive deficits, and lifespan. Prion-diseased mice were treated either intraperitoneally or intranasally with SB_NI_112. Behavioral and cognitive deficits were significantly protected by this combination of NF-κB and NLRP3 downregulators. Treatment reduced glial inflammation, protected against neuronal loss, prevented spongiotic change, rescued cognitive deficits, and significantly lengthened the lifespan of prion-diseased mice. We have identified a nontoxic, systemic pharmacologic that downregulates NF-κB and NLRP3, prevents neuronal death, and slows the progression of neurodegenerative diseases. Though mouse models do not always predict human patient success and the study was limited due to sample size and number of dosing methods utilized, these findings serve as a proof of principle for continued translation of the therapeutic SB_NI_112 for prion disease and other neurodegenerative diseases. Based on the success in a murine prion model, we will continue testing SB_NI_112 in a variety of neurodegenerative disease models, including Alzheimer's disease and Parkinson's disease.


Subject(s)
Alzheimer Disease , Neurodegenerative Diseases , Parkinson Disease , Prion Diseases , Prions , Proteostasis Deficiencies , Humans , Mice , Animals , Neurodegenerative Diseases/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , NF-kappa B/metabolism , Alzheimer Disease/metabolism , Neuroinflammatory Diseases , Down-Regulation , Parkinson Disease/metabolism , Neurons/metabolism , Prion Diseases/drug therapy , Prion Diseases/metabolism , Prions/metabolism , Inflammation/metabolism , Proteostasis Deficiencies/drug therapy , Proteostasis Deficiencies/metabolism
18.
Small ; 9(12): 2171-81, 2013 Jun 24.
Article in English | MEDLINE | ID: mdl-23322550

ABSTRACT

In biological environments, nanomaterials associate with proteins forming a protein corona (PC). The PC may alter the nanomaterial's pharmacokinetics and pharmacodynamics, thereby influencing toxicity. Using a label-free mass spectrometry-based proteomics approach, the composition of the PC is examined for a set of nanotubes (NTs) including unmodified and carboxylated single- (SWCNT) and multi-walled carbon nanotubes (MWCNT), polyvinylpyrrolidone (PVP)-coated MWCNT (MWCNT-PVP), and nanoclay. NTs are incubated for 1 h in simulated cell culture conditions, then washed, resuspended in PBS, and assessed by liquid chromatography-tandem mass spectrometry (LC-MS/MS) for their associated PC. To determine those attributes that influence PC formation, the NTs are extensively characterized. NTs had negative zeta potentials in water (SWCNT-COOH < MWCNT-COOH < unmodified NTs) while carboxylation increases their hydrodynamic sizes. All NTs are also found to associate a common subset of proteins including albumin, titin, and apolipoproteins. SWCNT-COOH and MWCNT-COOH are found to bind the greatest number of proteins (181 and 133 respectively) compared to unmodified NTs (<100), suggesting covalent binding to protein amines. Modified NTs bind a number of unique proteins compared to unmodified NTs, implying hydrogen bonding and electrostatic interactions are involved in PC formation. PVP-coating of MWCNT did not influence PC composition, further reinforcing the possibility of hydrogen bonding and electrostatic interactions. No relationships are found between PC composition and corresponding isoelectric point, hydropathy, or aliphatic index, implying minimal roles of hydrophobic interaction and pi-stacking.


Subject(s)
Nanotubes, Carbon/chemistry , Proteins/chemistry , Culture Media , Mass Spectrometry , Povidone/chemistry , Proteomics
19.
J Biochem Mol Toxicol ; 27(1): 50-5, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23129019

ABSTRACT

The global market for nanomaterial-based products is forecasted to reach 100 billion dollars per annum for 2011-2015. Extensive manufacturing and the use of engineered nanomaterials have raised concerns regarding their impact on biological response in living organisms and the environment at large. The fundamental properties of nanomaterials exhibit a complex dependence upon several factors such as their morphology, size, defects, and chemical stability. Therefore, it is exceedingly difficult to correlate their biological response with their intricate physicochemical properties. For example, varying toxic response may ensue due to different methods of nanomaterial preparation, dissimilar impurities, and defects. In this review, we surveyed the existing literature on the dependence of cytotoxicity on physicochemical properties. We found that ENM size, shape, defect density, physicochemical stability, and surface modification to be the main causes that elicit altered physiological response or cytotoxicity.


Subject(s)
Nanostructures/chemistry , Nanostructures/toxicity , Animals , Humans , Nanoparticles/chemistry , Nanoparticles/toxicity , Surface Properties
20.
Proc Natl Acad Sci U S A ; 107(12): 5652-7, 2010 Mar 23.
Article in English | MEDLINE | ID: mdl-20231466

ABSTRACT

Bone marrow stromal cells [BMSCs; also known as mesenchymal stem cells (MSCs)] effectively suppress inflammatory responses in acute graft-versus-host disease in humans and in a number of disease models in mice. Many of the studies concluded that BMSC-driven immunomodulation is mediated by the suppression of proinflammatory Th1 responses while rebalancing the Th1/Th2 ratio toward Th2. In this study, using a ragweed induced mouse asthma model, we studied if BMSCs could be beneficial in an allergic, Th2-dominant environment. When BMSCs were injected i.v. at the time of the antigen challenge, they protected the animals from the majority of asthma-specific pathological changes, including inhibition of eosinophil infiltration and excess mucus production in the lung, decreased levels of Th2 cytokines (IL-4, IL-5, and IL-13) in bronchial lavage, and lowered serum levels of Th2 immunoglobulins (IgG1 and IgE). To explore the mechanism of the effect we used BMSCs isolated from a variety of knockout mice, performed in vivo blocking of cytokines and studied the effect of asthmatic serum and bronchoalveolar lavage from ragweed challenged animals on the BMSCs in vitro. Our results suggest that IL-4 and/or IL-13 activate the STAT6 pathway in the BMSCs resulting in an increase of their TGF-beta production, which seems to mediate the beneficial effect, either alone, or together with regulatory T cells, some of which might be recruited by the BMSCs. These data suggest that, in addition to focusing on graft-versus-host disease and autoimmune diseases, allergic conditions--specifically therapy resistant asthma--might also be a likely target of the recently discovered cellular therapy approach using BMSCs.


Subject(s)
Asthma/immunology , Mesenchymal Stem Cells/immunology , Transforming Growth Factor beta/immunology , Ambrosia/adverse effects , Ambrosia/immunology , Animals , Asthma/etiology , Asthma/pathology , Asthma/therapy , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , Cytokines/deficiency , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Humans , Immunosuppression Therapy , In Vitro Techniques , Lung/immunology , Lung/pathology , Mesenchymal Stem Cell Transplantation , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Th2 Cells/immunology , Transplantation, Homologous , Transplantation, Isogeneic
SELECTION OF CITATIONS
SEARCH DETAIL