1
|
Hong X, Morgenlander WR, Nadeau K, Wang G, Frischmeyer-Guerrerio PA, Pearson C, Adams WG, Ji H, Larman HB, Wang X. Maternal exposure to per- and polyfluoroalkyl substances and epitope level antibody response to vaccines against measles and rubella in children from the Boston birth cohort. ENVIRONMENT INTERNATIONAL 2025; 198:109433. [PMID: 40215916 DOI: 10.1016/j.envint.2025.109433] [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: 12/24/2024] [Revised: 03/20/2025] [Accepted: 04/02/2025] [Indexed: 04/26/2025]
Abstract
BACKGROUND Previous studies suggest that per- and polyfluoroalkyl substances (PFAS) may act as immune suppressants. However, research about the impact of PFAS exposure on antibody responses to the measles, mumps, rubella (MMR) vaccine is limited and inconsistent. METHODS This report includes 748 mother-child pairs from the Boston Birth Cohort, with 8 PFAS compounds measured in maternal plasma shortly after delivery. IgG reactivities to measles and rubella were profiled in cord blood and venous blood plasma during early childhood, using Phage ImmunoPrecipitation Sequencing. Linear regression models were applied to assess the relationships between log2-transformed PFAS and IgG reactivities as measured by Viral Aggregate Reactivity score (VARscore, with inverse normal transformation) for measles and rubella. Quantile g-computation was applied to evaluate the PFAS mixture - VARscore associations. RESULTS The detection rate for 8 PFAS compounds ranged from 90 % to 100 % in maternal plasma. Maternal PFAS burden score (P = 0.01), but not individual PFAS compounds, was associated with lower VARscore for measles in cord blood. In 348 children after receiving the MMR vaccine, three maternal PFAS compounds (Me-PFOSA-AcOH, PFHpS and PFHxS) were significantly associated with lower measles VARscore (P < 0.05). Me-PFOSA-AcOH and PFHxS were significantly associated with higher risk of having low reactivity to measles defined as VARscore < 25th percentile. PFAS mixture analysis revealed a significant inverse association between quantile of the PFAS mixture and measles VARscore (P = 0.025) in children after vaccination, with PFHxS as an important contributor to this association. These inverse associations were more pronounced in Black children (compared to non-Black children) and in preterm children (compared to term children). In comparison, no associations were found for rubella VARscore. CONCLUSIONS This prospective birth cohort study provides suggestive evidence that maternal PFAS exposure is associated with a reduced immune response to the measles vaccine, especially, among Black or preterm children.
Collapse
Affiliation(s)
- Xiumei Hong
- Center on the Early Life Origins of Disease, Department of Population, Family and Reproductive Health, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21205, USA.
| | - William R Morgenlander
- Institute for Cell Engineering, Division of Immunology, Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Kari Nadeau
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Guoying Wang
- Center on the Early Life Origins of Disease, Department of Population, Family and Reproductive Health, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Pamela A Frischmeyer-Guerrerio
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Colleen Pearson
- Department of Pediatrics, Boston University School of Medicine and Boston Medical Center, Boston, MA 02118, USA
| | - William G Adams
- Department of Pediatrics, Boston University School of Medicine and Boston Medical Center, Boston, MA 02118, USA
| | - Hongkai Ji
- Department of Biostatistics, Johns Hopkins University Bloomberg School of Public Health, 615 N Wolfe St, Baltimore, MD 21205, USA
| | - H Benjamin Larman
- Institute for Cell Engineering, Division of Immunology, Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Xiaobin Wang
- Center on the Early Life Origins of Disease, Department of Population, Family and Reproductive Health, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21205, USA; Division of General Pediatrics & Adolescent Medicine, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
2
|
Bharal B, Ruchitha C, Kumar P, Pandey R, Rachamalla M, Niyogi S, Naidu R, Kaundal RK. Neurotoxicity of per- and polyfluoroalkyl substances: Evidence and future directions. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 955:176941. [PMID: 39454776 DOI: 10.1016/j.scitotenv.2024.176941] [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: 07/08/2024] [Revised: 09/28/2024] [Accepted: 10/12/2024] [Indexed: 10/28/2024]
Abstract
Per- and polyfluoroalkyl substances (PFAS) are synthetic chemicals widely used in various products, including food packaging, textiles, and firefighting foam, owing to their unique properties such as amphiphilicity and strong CF bonds. Despite their widespread use, concerns have arisen due to their resistance to degradation and propensity for bioaccumulation in both environmental and human systems. Emerging evidence suggests a potential link between PFAS exposure and neurotoxic effects, spanning cognitive deficits, neurodevelopmental disorders, and neurodegenerative diseases. This review comprehensively synthesizes current knowledge on PFAS neurotoxicity, drawing insights from epidemiological studies, animal experiments, and mechanistic investigations. PFAS, known for their lipophilic nature, tend to accumulate in lipid-rich tissues, including the brain, breaching biological barriers such as the blood-brain barrier (BBB). The accumulation of PFAS within the central nervous system (CNS) has been implicated in a spectrum of neurological maladies. Neurotoxicity induced by PFAS manifests through a multitude of direct and indirect mechanisms. A growing body of research associated PFAS exposure with BBB disruption, calcium dysregulation, neurotransmitter alterations, neuroinflammation, oxidative stress, and mitochondrial dysfunction, all contributing to neuronal impairment. Despite notable strides in research, significant lacunae persist, necessitating further exploration to elucidate the full spectrum of PFAS-mediated neurotoxicity. Prospective research endeavors should prioritize developing biomarkers, delineating sensitive exposure windows, and exploring mitigation strategies aimed at safeguarding neurological integrity within populations vulnerable to PFAS exposure.
Collapse
Affiliation(s)
- Bhagyashree Bharal
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research-Raebareli (NIPER-R), Transit Campus, Bijnor-Sisendi Road, Sarojini Nagar, Near CRPF Base Camp, Lucknow, UP 226002, India
| | - Chanda Ruchitha
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research-Raebareli (NIPER-R), Transit Campus, Bijnor-Sisendi Road, Sarojini Nagar, Near CRPF Base Camp, Lucknow, UP 226002, India
| | - Paarth Kumar
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research-Raebareli (NIPER-R), Transit Campus, Bijnor-Sisendi Road, Sarojini Nagar, Near CRPF Base Camp, Lucknow, UP 226002, India
| | - Rukmani Pandey
- Department of Psychiatry, Center for Molecular Biology and Genetics of Neurodegeneration, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States of America
| | - Mahesh Rachamalla
- Department of Biology, University of Saskatchewan, Saskatoon, SK S7N 5E2, Canada
| | - Som Niyogi
- Department of Biology, University of Saskatchewan, Saskatoon, SK S7N 5E2, Canada; Toxicology Centre, University of Saskatchewan, 44 Campus Drive, Saskatoon, SK S7N 5B3, Canada
| | - Ravi Naidu
- Global Centre for Environmental Remediation (GCER), University of Newcastle, Callaghan, NSW 2308, Australia; CRC for Contamination Assessment and Remediation of the Environment (CRC CARE), University of Newcastle, Callaghan, NSW 2308, Australia.
| | - Ravinder K Kaundal
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research-Raebareli (NIPER-R), Transit Campus, Bijnor-Sisendi Road, Sarojini Nagar, Near CRPF Base Camp, Lucknow, UP 226002, India; Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research-Raebareli (NIPER-R), Transit Campus, Bijnor-Sisendi Road, Sarojini Nagar, Near CRPF Base Camp, Lucknow, UP 226002, India.
| |
Collapse
|
3
|
Phelps DW, Connors AM, Ferrero G, DeWitt JC, Yoder JA. Per- and polyfluoroalkyl substances alter innate immune function: evidence and data gaps. J Immunotoxicol 2024; 21:2343362. [PMID: 38712868 PMCID: PMC11249028 DOI: 10.1080/1547691x.2024.2343362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 04/10/2024] [Indexed: 05/08/2024] Open
Abstract
Per- and polyfluoroalkyl substances (PFASs) are a large class of compounds used in a variety of processes and consumer products. Their unique chemical properties make them ubiquitous and persistent environmental contaminants while also making them economically viable and socially convenient. To date, several reviews have been published to synthesize information regarding the immunotoxic effects of PFASs on the adaptive immune system. However, these reviews often do not include data on the impact of these compounds on innate immunity. Here, current literature is reviewed to identify and incorporate data regarding the effects of PFASs on innate immunity in humans, experimental models, and wildlife. Known mechanisms by which PFASs modulate innate immune function are also reviewed, including disruption of cell signaling, metabolism, and tissue-level effects. For PFASs where innate immune data are available, results are equivocal, raising additional questions about common mechanisms or pathways of toxicity, but highlighting that the innate immune system within several species can be perturbed by exposure to PFASs. Recommendations are provided for future research to inform hazard identification, risk assessment, and risk management practices for PFASs to protect the immune systems of exposed organisms as well as environmental health.
Collapse
Affiliation(s)
- Drake W. Phelps
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, NC
- Center for Environmental and Health Effects of PFAS, North Carolina State University, Raleigh, NC
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC
| | - Ashley M. Connors
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, NC
- Center for Environmental and Health Effects of PFAS, North Carolina State University, Raleigh, NC
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC
- Toxicology Program, North Carolina State University, Raleigh, NC
- Genetics and Genomics Academy, North Carolina State University, Raleigh, NC
| | - Giuliano Ferrero
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, NC
- Center for Environmental and Health Effects of PFAS, North Carolina State University, Raleigh, NC
| | - Jamie C. DeWitt
- Center for Environmental and Health Effects of PFAS, North Carolina State University, Raleigh, NC
- Center for Human Health and the Environment, North Carolina State University, Raleigh, NC
- Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR
| | - Jeffrey A. Yoder
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, NC
- Center for Environmental and Health Effects of PFAS, North Carolina State University, Raleigh, NC
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC
- Toxicology Program, North Carolina State University, Raleigh, NC
- Genetics and Genomics Academy, North Carolina State University, Raleigh, NC
- Center for Human Health and the Environment, North Carolina State University, Raleigh, NC
| |
Collapse
|
4
|
Ren J, Zuo J, Yin B, Huang D, Wen R, Pei H, Liu J, Zhang Y, Zhu S, Zhen S, Ma Y. Flaxseed Oil Alleviates PFOS-Induced Liver Injury by Regulating Hepatic Cholesterol Metabolism. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:23465-23477. [PMID: 39392608 DOI: 10.1021/acs.jafc.4c04438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Perfluorooctanesulfonate (PFOS) is a widespread, persistent environmental pollutant that exerts apparent liver toxicity. Flaxseed oil (FO), a dietary oil rich in α-linolenic acid, has been demonstrated to possess a diverse array of health benefits. However, whether FO protects against PFOS-induced liver injury and its underlying mechanisms remain unclear. C57/BL6 mice were orally treated with different concentrations of FO alone or in combination with 10 mg/kg of PFOS for 28 consecutive days. Blood and liver tissues were collected for proteomic, histopathological, biochemical, immunohistochemical, and molecular examinations. Results demonstrated that FO supplementation reduced PFOS-induced liver injury, as evidenced by a decrease in histopathological changes, serum transaminase (ALT and AST) levels, levels of oxidative stress, and inflammatory cytokine (TNF-α, IL-1β, and IL-6) levels. Proteomic analyses showed that differentially expressed proteins were enriched in cholesterol metabolic pathways when comparing the PFOS group to the FO supplementation groups. The expression of cholesterol metabolism-related proteins was also subsequently measured, revealing that FO supplementation decreased the protein expressions of SREBP2, HMGCR, and LDLR while increasing the expression of CYP7A1. This study demonstrates that FO can alleviate PFOS-induced hepatotoxicity by regulating hepatic cholesterol metabolism, indicating that FO may serve as an effective dietary intervention for preventing liver injury caused by PFOS.
Collapse
Affiliation(s)
- Jingyi Ren
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Shijiazhuang 050017, China
| | - Jinshi Zuo
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Shijiazhuang 050017, China
| | - Bowen Yin
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Shijiazhuang 050017, China
| | - Dan Huang
- Undergraduate of College of Public Health, Hebei Medical University, Shijiazhuang 050017, China
| | - Rui Wen
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Shijiazhuang 050017, China
| | - Huanting Pei
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Shijiazhuang 050017, China
| | - Jiarui Liu
- Undergraduate of College of Basic Medicine, Hebei Medical University, Shijiazhuang 050017, China
| | - Yadong Zhang
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Shijiazhuang 050017, China
| | - Siqi Zhu
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Shijiazhuang 050017, China
| | - Shuman Zhen
- Department of Radiotherapy, Fourth Hospital of Hebei Medical University, Shijiazhuang 050017, China
| | - Yuxia Ma
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Shijiazhuang 050017, China
| |
Collapse
|
5
|
Walker KA, Rhodes ST, Liberman DA, Gore AC, Bell MR. Microglial responses to inflammatory challenge in adult rats altered by developmental exposure to polychlorinated biphenyls in a sex-specific manner. Neurotoxicology 2024; 104:95-115. [PMID: 39038526 PMCID: PMC11548868 DOI: 10.1016/j.neuro.2024.07.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 07/11/2024] [Accepted: 07/17/2024] [Indexed: 07/24/2024]
Abstract
Polychlorinated biphenyls are ubiquitous environmental contaminants linkedc with peripheral immune and neural dysfunction. Neuroimmune signaling is critical to brain development and later health; however, effects of PCBs on neuroimmune processes are largely undescribed. This study extends our previous work in neonatal or adolescent rats by investigating longer-term effects of perinatal PCB exposure on later neuroimmune responses to an inflammatory challenge in adulthood. Male and female Sprague-Dawley rats were exposed to a low-dose, environmentally relevant, mixture of PCBs (Aroclors 1242, 1248, and 1254, 1:1:1, 20 μg / kg dam BW per gestational day) or oil control during gestation and via lactation. Upon reaching adulthood, rats were given a mild inflammatory challenge with lipopolysaccharide (LPS, 50 μg / kg BW, ip) or saline control and then euthanized 3 hours later for gene expression analysis or 24 hours later for immunohistochemical labeling of Iba1+ microglia. PCB exposure did not alter gene expression or microglial morphology independently, but instead interacted with the LPS challenge in brain region- and sex-specific ways. In the female hypothalamus, PCB exposure blunted LPS responses of neuroimmune and neuromodulatory genes without changing microglial morphology. In the female prefrontal cortex, PCBs shifted Iba1+ cells from reactive to hyperramified morphology in response to LPS. Conversely, in the male hypothalamus, PCBs shifted cell phenotypes from hyperramified to reactive morphologies in response to LPS. The results highlight the potential for long-lasting effects of environmental contaminants that are differentially revealed over a lifetime, sometimes only after a secondary challenge. These neuroimmune endpoints are possible mechanisms for PCB effects on a range of neural dysfunction in adulthood, including mental health and neurodegenerative disorders. The findings suggest possible interactions with other environmental challenges that also influence neuroimmune systems.
Collapse
Affiliation(s)
- Katherine A Walker
- Departments of Biological Sciences and Health Sciences, DePaul University, Chicago, IL 60614, USA.
| | - Simone T Rhodes
- Departments of Biological Sciences and Health Sciences, DePaul University, Chicago, IL 60614, USA.
| | - Deborah A Liberman
- Departments of Biological Sciences and Health Sciences, DePaul University, Chicago, IL 60614, USA.
| | - Andrea C Gore
- Division of Pharmacology and Toxicology, College of Pharmacy and Department of Psychology, University of Texas at Austin, Austin, TX 78712, USA.
| | - Margaret R Bell
- Departments of Biological Sciences and Health Sciences, DePaul University, Chicago, IL 60614, USA; Division of Pharmacology and Toxicology, College of Pharmacy and Department of Psychology, University of Texas at Austin, Austin, TX 78712, USA.
| |
Collapse
|
6
|
Sigvaldsen A, Højsager FD, Paarup HM, Beck IH, Timmermann CAG, Boye H, Nielsen F, Halldorsson TI, Nielsen C, Möller S, Barington T, Grandjean P, Jensen TK. Early-life exposure to perfluoroalkyl substances and serum antibody concentrations towards common childhood vaccines in 18-month-old children in the Odense Child Cohort. ENVIRONMENTAL RESEARCH 2024; 242:117814. [PMID: 38042520 DOI: 10.1016/j.envres.2023.117814] [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: 06/12/2023] [Revised: 11/23/2023] [Accepted: 11/27/2023] [Indexed: 12/04/2023]
Abstract
Exposure to per- and polyfluoroalkyl substances (PFAS) has been associated with reduced antibody response to childhood vaccinations. Previous studies have mostly focused on antibodies against diphtheria or tetanus, while fewer studies have assessed antibodies toward attenuated viruses, such as measles, mumps or rubella (MMR). Therefore, we set out to determine associations between prenatal and early postnatal PFAS exposure and vaccine-specific Immunoglobulin G (IgG) in the background-exposed Odense Child Cohort. Blood samples were drawn in pregnancy at gestation weeks 8-16 and from the offspring at age 18 months. In the maternal serum samples we quantified perfluorooctane sulfonic acid (PFOS), perfluorooctanoic acid (PFOA), perfluorohexane sulfonic acid (PFHxS), perfluorononanoic acid (PFNA) and perfluorodecanoic acid (PFDA). In the offspring serum samples we quantified the same five PFAS compounds and IgG towards diphtheria, tetanus and MMR. A total of 880 and 841 children were included in the analyses of diphtheria and tetanus or MMR, respectively. Multiple linear regression models were used for estimation of difference in virus-specific IgG per doubling of PFAS concentrations. Maternal PFAS concentrations were non-significantly inversely associated with most vaccine-specific antibody concentrations. Likewise, child PFAS concentrations were associated with non-significant reductions of antibodies towards tetanus and MMR. A significant reduction in the percent difference in mumps antibody concentration per doubling of child PFNA (-9.2% (95% confidence interval: -17.4;-0.2)), PFHxS (-8.3% (-15.0;-1.0) and PFOS (-7.9% (-14.8;-0.4) was found. These findings are of public health concern, as inadequate response towards childhood vaccines may represent a more general immune dysfunction.
Collapse
Affiliation(s)
- Annika Sigvaldsen
- Department of Clinical Pharmacology, Pharmacy and Environmental Medicine, Institute of Public Health, University of Southern Denmark, J.B. Winsløwsvej 17A, 5000, Odense, Denmark; Hans Christian Andersen Children's Hospital, Odense University Hospital, Odense, Denmark.
| | - Frederik Damsgaard Højsager
- Department of Clinical Pharmacology, Pharmacy and Environmental Medicine, Institute of Public Health, University of Southern Denmark, J.B. Winsløwsvej 17A, 5000, Odense, Denmark; Hans Christian Andersen Children's Hospital, Odense University Hospital, Odense, Denmark
| | | | - Iben Have Beck
- Department of Clinical Pharmacology, Pharmacy and Environmental Medicine, Institute of Public Health, University of Southern Denmark, J.B. Winsløwsvej 17A, 5000, Odense, Denmark; Hans Christian Andersen Children's Hospital, Odense University Hospital, Odense, Denmark
| | | | - Henriette Boye
- Hans Christian Andersen Children's Hospital, Odense University Hospital, Odense, Denmark; Odense Child Cohort, Odense University Hospital, Odense, Denmark
| | - Flemming Nielsen
- Department of Clinical Pharmacology, Pharmacy and Environmental Medicine, Institute of Public Health, University of Southern Denmark, J.B. Winsløwsvej 17A, 5000, Odense, Denmark
| | | | - Christel Nielsen
- Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Sören Möller
- Open Patient data Explorative Network, Odense University Hospital, Odense, Denmark; Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Torben Barington
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark; Department of Clinical Immunology, Odense University Hospital, Odense, Denmark
| | - Philippe Grandjean
- Department of Clinical Pharmacology, Pharmacy and Environmental Medicine, Institute of Public Health, University of Southern Denmark, J.B. Winsløwsvej 17A, 5000, Odense, Denmark; Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, RI, USA
| | - Tina Kold Jensen
- Department of Clinical Pharmacology, Pharmacy and Environmental Medicine, Institute of Public Health, University of Southern Denmark, J.B. Winsløwsvej 17A, 5000, Odense, Denmark; Hans Christian Andersen Children's Hospital, Odense University Hospital, Odense, Denmark
| |
Collapse
|
7
|
Li S, Gao Z, Zhong M, Bi H, Li J. Identification of the mechanisms underlying per- and polyfluoroalkyl substance-induced hippocampal neurotoxicity as determined by network pharmacology and molecular docking analyses. Toxicol Res (Camb) 2023; 12:1126-1134. [PMID: 38145100 PMCID: PMC10734622 DOI: 10.1093/toxres/tfad104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 09/18/2023] [Accepted: 10/09/2023] [Indexed: 12/26/2023] Open
Abstract
Background Per- and polyfluoroalkyl substances (PFASs) are a class of environmental contaminants that pose significant health risks to both animals and humans. Although the hippocampal neurotoxic effects of numerous PFASs have been reported, the underlying mechanisms of combined exposure to PFASs-induced hippocampal neurotoxicity remain unclear. Methods In this study, network pharmacology analysis was performed to identify the intersectional targets of PFASs for possible associations with hippocampal neurotoxicity. The evaluation of the influence of PFASs on intersectional targets was assessed using a weighted method. Additionally, Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis of the screened targets were performed, the intersected hub targets calculated by various algorithms were screened in the network and molecular docking was also used to analyze binding activities. Results Our results indicated that eight PFASs, which acted on key targets (MYC, ESR1, STAT3, RELA, MAPK3) impacted the NF-κB signaling pathway, STAT3 signaling pathway, and MAPK signaling pathways to exert neurotoxicity in the hippocampus. The molecular docking results revealed that PFASs have strong binding potential to the hub targets. Conclusions Our findings provided a basis for future studies to investigate the detailed mechanisms of PFASs-induced hippocampal neurotoxicity and to develop preventative and control strategies.
Collapse
Affiliation(s)
- Shirui Li
- Department of Biostatistics, College of Public Health, Xuzhou Medical University, 209 Tongshan Road, Yun Long District, Xuzhou 221000, China
| | - Zhihui Gao
- Department of Biostatistics, College of Public Health, Xuzhou Medical University, 209 Tongshan Road, Yun Long District, Xuzhou 221000, China
| | - Meihan Zhong
- Department of Biostatistics, College of Public Health, Xuzhou Medical University, 209 Tongshan Road, Yun Long District, Xuzhou 221000, China
| | - Haoran Bi
- Department of Biostatistics, College of Public Health, Xuzhou Medical University, 209 Tongshan Road, Yun Long District, Xuzhou 221000, China
| | - Jianan Li
- Department of Occupational and Environmental Health, College of Public Health, Xuzhou Medical University, 209 Tongshan Road, Yun Long District, Xuzhou 221000, China
| |
Collapse
|
8
|
Tan Y, Taibl KR, Dunlop AL, Barr DB, Panuwet P, Yakimavets V, Kannan K, Corwin EJ, Ryan PB, Eatman JA, Liang D, Eick SM. Association between a Mixture of Per- and Polyfluoroalkyl Substances (PFAS) and Inflammatory Biomarkers in the Atlanta African American Maternal-Child Cohort. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2023; 57:13419-13428. [PMID: 37649345 PMCID: PMC10900195 DOI: 10.1021/acs.est.3c04688] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
Abstract
Per- and polyfluoroalkyl substances (PFAS) have been identified as environmental contributors to adverse birth outcomes. One potential mechanistic pathway could be through PFAS-related inflammation and cytokine production. Here, we examined associations between a PFAS mixture and inflammatory biomarkers during early and late pregnancy from participants enrolled in the Atlanta African American Maternal-Child Cohort (N = 425). Serum concentrations of multiple PFAS were detected in >90% samples at 8-14 weeks gestation. Serum concentrations of interferon-γ (IFN-γ), interleukin 6 (IL-6), interleukin 10 (IL-10), tumor necrosis factor-α (TNF-α), and C-reactive protein (CRP) were measured at up to two time points (8-14 weeks and 24-30 weeks gestation). The effect of the PFAS mixture on each inflammatory biomarker was examined using quantile g-computation, Bayesian kernel machine regression (BKMR), Bayesian Weighted Sums (BWS), and weighted quantile sum (WQS) regression. Across all models, the PFAS mixture was associated with increased IFN-γ, IL-10, and TNF-α at both time points, with the strongest effects being observed at 24-30 weeks. Using quantile g-computation, increasing concentrations of a PFAS mixture were associated with a 29% (95% confidence interval = 18.0%, 40.7%) increase in TNF-α at 24-30 weeks. Similarly, using BWS, the PFAS mixture was associated with increased TNF-α at 24-30 weeks (summed effect = 0.29, 95% highest posterior density = 0.17, 0.41). The PFAS mixture was also positively associated with TNF-α at 24-30 weeks using BKMR [75th vs 50th percentile: 17.1% (95% credible interval = 7.7%, 27.4%)]. Meanwhile, PFOS was consistently the main drivers of overall mixture effect across four methods. Our findings indicated an increase in prenatal PFAS exposure is associated with an increase in multiple pro-inflammatory cytokines, potentially contributing to adverse pregnancy outcomes.
Collapse
Affiliation(s)
- Youran Tan
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, 30329, USA
| | - Kaitlin R. Taibl
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, 30329, USA
| | - Anne L. Dunlop
- Department of Gynecology and Obstetrics, School of Medicine, Emory University, Atlanta, GA, 30329, USA
| | - Dana Boyd Barr
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, 30329, USA
| | - Parinya Panuwet
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, 30329, USA
| | - Volha Yakimavets
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, 30329, USA
| | - Kurunthachalam Kannan
- Department of Pediatrics, New York University School of Medicine, New York, NY, 10016, USA
- Department of Environmental Medicine, New York University School of Medicine, New York, 10016, NY, USA
| | | | - P. Barry Ryan
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, 30329, USA
| | - Jasmin A. Eatman
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, 30329, USA
| | - Donghai Liang
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, 30329, USA
| | - Stephanie M. Eick
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, 30329, USA
| |
Collapse
|
9
|
Guan D, Li Y, Cui Y, Zhao H, Dong N, Wang K, Ren D, Song T, Wang X, Jin S, Gao Y, Wang M. 5-HMF attenuates inflammation and demyelination in experimental autoimmune encephalomyelitis mice by inhibiting the MIF-CD74 interaction. Acta Biochim Biophys Sin (Shanghai) 2023; 55:1222-1233. [PMID: 37431183 PMCID: PMC10448060 DOI: 10.3724/abbs.2023105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Accepted: 02/10/2023] [Indexed: 07/12/2023] Open
Abstract
The neuroprotective role of 5-hydroxymethyl-2-furfural (5-HMF) has been demonstrated in a variety of neurological diseases. The aim of this study is to investigate the effect of 5-HMF on multiple sclerosis (MS). IFN-γ-stimulated murine microglia (BV2 cells) are considered a cell model of MS. With 5-HMF treatment, microglial M1/2 polarization and cytokine levels are detected. The interaction of 5-HMF with migration inhibitory factor (MIF) is predicted using online databases. The experimental autoimmune encephalomyelitis (EAE) mouse model is established, followed by a 5-HMF injection. The results show that 5-HMF facilitates IFN-γ-stimulated microglial M2 polarization and attenuates the inflammatory response. According to the network pharmacology and molecular docking results, 5-HMF has a binding site for MIF. Further results show that blocking MIF activity or silencing CD74 enhances microglial M2 polarization, reduces inflammatory activity, and prevents ERK1/2 phosphorylation. 5-HMF inhibits the MIF-CD74 interaction by binding to MIF, thereby inhibiting microglial M1 polarization and enhancing the anti-inflammatory response. 5-HMF ameliorates EAE, inflammation, and demyelination in vivo. In conclusion, our research indicates that 5-HMF promotes microglial M2 polarization by inhibiting the MIF-CD74 interaction, thereby attenuating inflammation and demyelination in EAE mice.
Collapse
Affiliation(s)
- Dongsheng Guan
- Department of Neurologythe Second Clinical Medical CollegeHenan University of Traditional Chinese MedicineZhengzhou450002China
| | - Yingxia Li
- The College of Basic MedicineHenan University of Traditional Chinese MedicineZhengzhou450046China
| | - Yinglin Cui
- Department of Neurologythe Second Clinical Medical CollegeHenan University of Traditional Chinese MedicineZhengzhou450002China
| | - Huanghong Zhao
- Department of Neurologythe Second Clinical Medical CollegeHenan University of Traditional Chinese MedicineZhengzhou450002China
| | - Ning Dong
- Department of Neurologythe Second Clinical Medical CollegeHenan University of Traditional Chinese MedicineZhengzhou450002China
| | - Kun Wang
- Department of Pharmacythe Second Clinical Medical CollegeHenan University of Traditional Chinese MedicineZhengzhou450002China
| | - Deqi Ren
- Department of Neurologythe Second Clinical Medical CollegeHenan University of Traditional Chinese MedicineZhengzhou450002China
| | - Tiantian Song
- Department of Neurologythe Second Clinical Medical CollegeHenan University of Traditional Chinese MedicineZhengzhou450002China
| | - Xiaojing Wang
- Department of Neurologythe Second Clinical Medical CollegeHenan University of Traditional Chinese MedicineZhengzhou450002China
| | - Shijie Jin
- Department of Neurologythe Second Clinical Medical CollegeHenan University of Traditional Chinese MedicineZhengzhou450002China
| | - Yinghe Gao
- Department of Neurologythe Second Clinical Medical CollegeHenan University of Traditional Chinese MedicineZhengzhou450002China
| | - Mengmeng Wang
- Department of Neurologythe Second Clinical Medical CollegeHenan University of Traditional Chinese MedicineZhengzhou450002China
| |
Collapse
|
10
|
Nagar N, Saxena H, Pathak A, Mishra A, Poluri KM. A review on structural mechanisms of protein-persistent organic pollutant (POP) interactions. CHEMOSPHERE 2023; 332:138877. [PMID: 37164191 DOI: 10.1016/j.chemosphere.2023.138877] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 04/20/2023] [Accepted: 05/06/2023] [Indexed: 05/12/2023]
Abstract
With the advent of the industrial revolution, the accumulation of persistent organic pollutants (POPs) in the environment has become ubiquitous. POPs are halogen-containing organic molecules that accumulate, and remain in the environment for a long time, thus causing toxic effects in living organisms. POPs exhibit a high affinity towards biological macromolecules such as nucleic acids, proteins and lipids, causing genotoxicity and impairment of homeostasis in living organisms. Proteins are essential members of the biological assembly, as they stipulate all necessary processes for the survival of an organism. Owing to their stereochemical features, POPs and their metabolites form energetically favourable complexes with proteins, as supported by biological and dose-dependent toxicological studies. Although individual studies have reported the biological aspects of protein-POP interactions, no comprehensive study summarizing the structural mechanisms, thermodynamics and kinetics of protein-POP complexes is available. The current review identifies and classifies protein-POP interaction according to the structural and functional basis of proteins into five major protein targets, including digestive and other enzymes, serum proteins, transcription factors, transporters, and G-protein coupled receptors. Further, analysis detailing the molecular interactions and structural mechanism evidenced that H-bonds, van der Waals, and hydrophobic interactions essentially mediate the formation of protein-POP complexes. Moreover, interaction of POPs alters the protein conformation through kinetic and thermodynamic processes like competitive inhibition and allostery to modulate the cellular signalling processes, resulting in various pathological conditions such as cancers and inflammations. In summary, the review provides a comprehensive insight into the critical structural/molecular aspects of protein-POP interactions.
Collapse
Affiliation(s)
- Nupur Nagar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, 247667, Uttarakhand, India
| | - Harshi Saxena
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, 247667, Uttarakhand, India
| | - Aakanksha Pathak
- Centre for Nanotechnology, Indian Institute of Technology Roorkee, Roorkee, 247667, Uttarakhand, India
| | - Amit Mishra
- Cellular and Molecular Neurobiology Unit, Indian Institute of Technology Jodhpur, Jodhpur, 342011, Rajasthan, India
| | - Krishna Mohan Poluri
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, 247667, Uttarakhand, India; Centre for Nanotechnology, Indian Institute of Technology Roorkee, Roorkee, 247667, Uttarakhand, India.
| |
Collapse
|
11
|
Ehrlich V, Bil W, Vandebriel R, Granum B, Luijten M, Lindeman B, Grandjean P, Kaiser AM, Hauzenberger I, Hartmann C, Gundacker C, Uhl M. Consideration of pathways for immunotoxicity of per- and polyfluoroalkyl substances (PFAS). Environ Health 2023; 22:19. [PMID: 36814257 PMCID: PMC9944481 DOI: 10.1186/s12940-022-00958-5] [Citation(s) in RCA: 90] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 12/30/2022] [Indexed: 05/02/2023]
Abstract
BACKGROUND Per- and polyfluoroalkyl substances (PFAS) are of public health concern, because of their ubiquitous and extremely persistent occurrence, and depending on their structure, their bio-accumulative, mobile and toxic properties. Human health effects associated with exposure to PFAS include adverse effects on the immune system. In 2020, EFSA (the European Food Safety Authority) defined adverse effects on the immune system as the most critical effect for human health risk assessment, based on reduced antibody responses to childhood vaccines and similar effects observed in experimental animal studies. Likewise, the U.S. EPA (Environmental Protection Agency) considers PFAS-induced immunotoxicity, especially in children, as the critical effect for risk assessment. However, the mechanisms by which antibody concentrations are impacted are not completely understood. Furthermore, other targets of the immune system functions have been reported in the literature. OBJECTIVE The aim of this review is to explore PFAS-associated immune-related effects. This includes, relevant mechanisms that may underlie the observed effects on the immune system, immunosuppression as well as immunoenhancement, such as i) modulation of cell signalling and nuclear receptors, such as NF-κB and PPARs; ii) alteration of calcium signalling and homoeostasis in immune cells; iii) modulation of immune cell populations; iv) oxidative stress and v) impact on fatty acid metabolism & secondary effects on the immune system. METHODS A literature research was conducted using three databases (Web of Science, PubMed, and Scopus), which were searched in July 2021 for relevant studies published in the time frame from 2018 to 2021. In total, 487 publications were identified as potentially eligible and following expert-based judgement, articles relevant for mechanisms of PFAS induced immunotoxicity are discussed. CONCLUSIONS Taken together, we show that there is substantial evidence from both in vitro and in vivo experimental as well as epidemiological studies, supporting that various PFAS, not only PFOA and PFOS, affect multiple aspects of the immune system. Timing of exposure is critical, because the developing immune system is especially vulnerable to toxic insults, resulting in a higher risk of particularly adverse immune effects but also other organs later in life.
Collapse
Affiliation(s)
- Veronika Ehrlich
- Environment Agency Austria (Umweltbundesamt GmbH), Spittelauer Lände 5, 1090, Vienna, Austria
| | - Wieneke Bil
- Centre for Safety of Substances and Products, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Rob Vandebriel
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Berit Granum
- Division of Climate and Environment Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Mirjam Luijten
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Birgitte Lindeman
- Division of Climate and Environment Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Philippe Grandjean
- Department of Public Health, University of Southern Denmark, Odense, Denmark
- Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, RI, USA
| | - Andreas-Marius Kaiser
- Environment Agency Austria (Umweltbundesamt GmbH), Spittelauer Lände 5, 1090, Vienna, Austria
| | - Ingrid Hauzenberger
- Environment Agency Austria (Umweltbundesamt GmbH), Spittelauer Lände 5, 1090, Vienna, Austria
| | - Christina Hartmann
- Environment Agency Austria (Umweltbundesamt GmbH), Spittelauer Lände 5, 1090, Vienna, Austria
| | - Claudia Gundacker
- Center for Pathobiochemistry and Genetics, Medical University of Vienna, Vienna, Austria
| | - Maria Uhl
- Environment Agency Austria (Umweltbundesamt GmbH), Spittelauer Lände 5, 1090, Vienna, Austria.
| |
Collapse
|
12
|
Wang D, Tan Z, Yang J, Li L, Li H, Zhang H, Liu H, Liu Y, Wang L, Li Q, Guo H. Perfluorooctane sulfonate promotes atherosclerosis by modulating M1 polarization of macrophages through the NF-κB pathway. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 249:114384. [PMID: 36512850 DOI: 10.1016/j.ecoenv.2022.114384] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 11/27/2022] [Accepted: 12/01/2022] [Indexed: 06/17/2023]
Abstract
Perfluorooctane sulfonate (PFOS) is a widely used and distributed perfluorinated compounds and is reported to be harmful to cardiovascular health; however, the direct association between PFOS exposure and atherosclerosis and the underlying mechanisms remain unknown. Therefore, this study aimed to investigate the effects of PFOS exposure on the atherosclerosis progression and the underlying mechanisms. PFOS was administered through oral gavage to apolipoprotein E-deficient (ApoE-/-) mice for 12 weeks. PFOS exposure significantly increased pulse wave velocity (PWV) and intima-media thickness (IMT), increased aortic plaque burden and vulnerability, and elevated serum lipid and inflammatory cytokine levels. PFOS promoted aortic and RAW264.7 M1 macrophage polarization, which increased the secretion of nitric oxide synthase (iNOS) and pro-inflammatory factors (tumor necrosis factor-α [TNF-α], interleukin-6 [IL-6], and interleukin-1β [IL-1β]), and suppressed M2 macrophage polarization, which decreased the expression of CD206, arginine I (Arg-1), and interleukin-10 (IL-10). Moreover, PFOS activated nuclear factor-kappa B (NF-κB) in the aorta and macrophages. BAY11-7082 was used to inhibit NF-κB-alleviated M1 macrophage polarization and the inflammatory response induced by PFOS in RAW264.7 macrophages. Our results are the first to reveal the acceleratory effect of PFOS on the atherosclerosis progression in ApoE-/- mice, which is associated with the NF-κB activation of macrophages to M1 polarization to induce inflammation.
Collapse
Affiliation(s)
- Dan Wang
- Department of Toxicology, Hebei Medical University, Shijiazhuang, China
| | - Zhenzhen Tan
- Department of Toxicology, Hebei Medical University, Shijiazhuang, China
| | - Jing Yang
- Department of Toxicology, Hebei Medical University, Shijiazhuang, China
| | - Longfei Li
- Department of Toxicology, Hebei Medical University, Shijiazhuang, China
| | - Haoran Li
- Department of Toxicology, Hebei Medical University, Shijiazhuang, China
| | - Huaxing Zhang
- Core Facilities and Centers, Hebei Medical University, Shijiazhuang, China
| | - Heqiong Liu
- Department of Toxicology, Hebei Medical University, Shijiazhuang, China
| | - Yi Liu
- Department of Toxicology, Hebei Medical University, Shijiazhuang, China
| | - Lei Wang
- Department of Medicinal Chemistry, Hebei Medical University, Shijiazhuang, China
| | - Qian Li
- Department of Physiology, Hebei Medical University, Shijiazhuang, China.
| | - Huicai Guo
- Department of Toxicology, Hebei Medical University, Shijiazhuang, China; Hebei Key Laboratory of Environment and Human Health, Shijiazhuang, China.
| |
Collapse
|
13
|
Characterization of the RAS/RAF/ERK Signal Cascade as a Novel Regulating Factor in Alpha-Amanitin-Induced Cytotoxicity in Huh-7 Cells. Int J Mol Sci 2022; 23:ijms232012294. [PMID: 36293151 PMCID: PMC9603094 DOI: 10.3390/ijms232012294] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 10/05/2022] [Accepted: 10/10/2022] [Indexed: 12/24/2022] Open
Abstract
The well-known hepatotoxicity mechanism resulting from alpha-amanitin (α-AMA) exposure arises from RNA polymerase II (RNAP II) inhibition. RNAP Ⅱ inhibition occurs through the dysregulation of mRNA synthesis. However, the signaling pathways in hepatocytes that arise from α-AMA have not yet been fully elucidated. Here, we identified that the RAS/RAF/ERK signaling pathway was activated through quantitative phosphoproteomic and molecular biological analyses in Huh-7 cells. Bioinformatics analysis showed that α-AMA exposure increased protein phosphorylation in a time-dependent α-AMA exposure. In addition, phosphorylation increased not only the components of the ERK signaling pathway but also U2AF65 and SPF45, known splicing factors. Therefore, we propose a novel mechanism of α-AMA as follows. The RAS/RAF/ERK signaling pathway involved in aberrant splicing events is activated by α-AMA exposure followed by aberrant splicing events leading to cell death in Huh-7 cells.
Collapse
|
14
|
Clerbaux LA, Albertini MC, Amigó N, Beronius A, Bezemer GFG, Coecke S, Daskalopoulos EP, del Giudice G, Greco D, Grenga L, Mantovani A, Muñoz A, Omeragic E, Parissis N, Petrillo M, Saarimäki LA, Soares H, Sullivan K, Landesmann B. Factors Modulating COVID-19: A Mechanistic Understanding Based on the Adverse Outcome Pathway Framework. J Clin Med 2022; 11:4464. [PMID: 35956081 PMCID: PMC9369763 DOI: 10.3390/jcm11154464] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 07/05/2022] [Accepted: 07/06/2022] [Indexed: 12/10/2022] Open
Abstract
Addressing factors modulating COVID-19 is crucial since abundant clinical evidence shows that outcomes are markedly heterogeneous between patients. This requires identifying the factors and understanding how they mechanistically influence COVID-19. Here, we describe how eleven selected factors (age, sex, genetic factors, lipid disorders, heart failure, gut dysbiosis, diet, vitamin D deficiency, air pollution and exposure to chemicals) influence COVID-19 by applying the Adverse Outcome Pathway (AOP), which is well-established in regulatory toxicology. This framework aims to model the sequence of events leading to an adverse health outcome. Several linear AOPs depicting pathways from the binding of the virus to ACE2 up to clinical outcomes observed in COVID-19 have been developed and integrated into a network offering a unique overview of the mechanisms underlying the disease. As SARS-CoV-2 infectibility and ACE2 activity are the major starting points and inflammatory response is central in the development of COVID-19, we evaluated how those eleven intrinsic and extrinsic factors modulate those processes impacting clinical outcomes. Applying this AOP-aligned approach enables the identification of current knowledge gaps orientating for further research and allows to propose biomarkers to identify of high-risk patients. This approach also facilitates expertise synergy from different disciplines to address public health issues.
Collapse
Affiliation(s)
- Laure-Alix Clerbaux
- European Commission, Joint Research Centre (JRC), 21027 Ispra, Italy; (S.C.); (E.P.D.); (N.P.); (M.P.); (B.L.)
| | | | - Núria Amigó
- Biosfer Teslab SL., 43204 Reus, Spain;
- Department of Basic Medical Sciences, Universitat Rovira i Virgili (URV), 23204 Reus, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
| | - Anna Beronius
- Institute of Environmental Medicine, Karolinska Institutet, 17177 Stockholm, Sweden;
| | - Gillina F. G. Bezemer
- Impact Station, 1223 JR Hilversum, The Netherlands;
- Department of Pharmaceutical Sciences, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands
| | - Sandra Coecke
- European Commission, Joint Research Centre (JRC), 21027 Ispra, Italy; (S.C.); (E.P.D.); (N.P.); (M.P.); (B.L.)
| | - Evangelos P. Daskalopoulos
- European Commission, Joint Research Centre (JRC), 21027 Ispra, Italy; (S.C.); (E.P.D.); (N.P.); (M.P.); (B.L.)
| | - Giusy del Giudice
- Finnish Hub for Development and Validation of Integrated Approaches (FHAIVE), Faculty of Medicine and Health Technology, Tampere University, 33100 Tampere, Finland; (G.d.G.); (D.G.); (L.A.S.)
| | - Dario Greco
- Finnish Hub for Development and Validation of Integrated Approaches (FHAIVE), Faculty of Medicine and Health Technology, Tampere University, 33100 Tampere, Finland; (G.d.G.); (D.G.); (L.A.S.)
| | - Lucia Grenga
- Département Médicaments et Technologies pour la Santé (DMTS), Université Paris-Saclay, CEA, INRAE, SPI, F-30200 Bagnols-sur-Ceze, France;
| | - Alberto Mantovani
- Department of Food Safety, Nutrition and Veterinary Public Health, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Amalia Muñoz
- European Commission, Joint Research Centre (JRC), 2440 Geel, Belgium;
| | - Elma Omeragic
- Faculty of Pharmacy, University of Sarajevo, 71000 Sarajevo, Bosnia and Herzegovina;
| | - Nikolaos Parissis
- European Commission, Joint Research Centre (JRC), 21027 Ispra, Italy; (S.C.); (E.P.D.); (N.P.); (M.P.); (B.L.)
| | - Mauro Petrillo
- European Commission, Joint Research Centre (JRC), 21027 Ispra, Italy; (S.C.); (E.P.D.); (N.P.); (M.P.); (B.L.)
| | - Laura A. Saarimäki
- Finnish Hub for Development and Validation of Integrated Approaches (FHAIVE), Faculty of Medicine and Health Technology, Tampere University, 33100 Tampere, Finland; (G.d.G.); (D.G.); (L.A.S.)
| | - Helena Soares
- Laboratory of Immunobiology and Pathogenesis, Chronic Diseases Research Centre, Faculdade de Ciências Médicas Medical School, University of Lisbon, 1649-004 Lisbon, Portugal;
| | - Kristie Sullivan
- Physicians Committee for Responsible Medicine, Washington, DC 20016, USA;
| | - Brigitte Landesmann
- European Commission, Joint Research Centre (JRC), 21027 Ispra, Italy; (S.C.); (E.P.D.); (N.P.); (M.P.); (B.L.)
| |
Collapse
|
15
|
Wang P, Liu D, Yan S, Cui J, Liang Y, Ren S. Adverse Effects of Perfluorooctane Sulfonate on the Liver and Relevant Mechanisms. TOXICS 2022; 10:toxics10050265. [PMID: 35622678 PMCID: PMC9144769 DOI: 10.3390/toxics10050265] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 05/13/2022] [Accepted: 05/17/2022] [Indexed: 02/07/2023]
Abstract
Perfluorooctane sulfonate (PFOS) is a persistent, widely present organic pollutant. PFOS can enter the human body through drinking water, ingestion of food, contact with utensils containing PFOS, and occupational exposure to PFOS, and can have adverse effects on human health. Increasing research shows that the liver is the major target of PFOS, and that PFOS can damage liver tissue and disrupt its function; however, the exact mechanisms remain unclear. In this study, we reviewed the adverse effects of PFOS on liver tissue and cells, as well as on liver function, to provide a reference for subsequent studies related to the toxicity of PFOS and liver injury caused by PFOS.
Collapse
|
16
|
Liang L, Pan Y, Bin L, Liu Y, Huang W, Li R, Lai KP. Immunotoxicity mechanisms of perfluorinated compounds PFOA and PFOS. CHEMOSPHERE 2022; 291:132892. [PMID: 34780734 DOI: 10.1016/j.chemosphere.2021.132892] [Citation(s) in RCA: 76] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 11/09/2021] [Accepted: 11/11/2021] [Indexed: 05/14/2023]
Abstract
Perfluorinated and polyfluorinated compounds (PFASs) are a class of synthetic chemical substances that are widely used in human production and life, such as fire-fighting foams, textiles and clothing, surfactants, and surface protective agents. Perfluorooctane sulfonic acid (PFOS) and perfluorooctanoic acid (PFOA) are the most abundant and common perfluorinated compounds in biota and humans. Currently, PFOA and PFOS have been listed in the Stockholm Convention on Persistent Organic Pollutants, and their production has been halted in many countries. However, because the high-energy carbon-fluorine bond can make it resistant to hydrolysis, photolysis, microbial degradation, and vertebrate metabolism, PFOA and PFOS show environmental persistence and bioaccumulation and hence, are of great concern to humans and wildlife. PFOA and PFOS have toxic effects on the immune system of the body. This article reviewed the effects of PFOA and PFOS on immune organs such as the spleen, bone marrow, and thymus of mice and zebrafish, and the effects on non-specific immune functions such as the skin barrier, intestinal mucosal barrier, and humoral immunity. We also reviewed the influence of specific immune functions based on cellular immunity, and further summarized the possible immune toxicity mechanisms such as AIM2 inflammasome activation, gene dysregulation, and signal pathway disorders caused by PFOA and PFOS. The aim of this review was to provide a reference for further understanding of the immunotoxicity and the responsible mechanism of PFOA and PFOS.
Collapse
Affiliation(s)
- Luyun Liang
- School of Lingui Clinical Medicine, Guilin Medical University, Guilin, PR China; Laboratory of Environmental Pollution and Integrative Omics, Guilin Medical University, Guilin, PR China
| | - Yongling Pan
- School of Lingui Clinical Medicine, Guilin Medical University, Guilin, PR China; Laboratory of Environmental Pollution and Integrative Omics, Guilin Medical University, Guilin, PR China
| | - Lihua Bin
- School of Lingui Clinical Medicine, Guilin Medical University, Guilin, PR China; Laboratory of Environmental Pollution and Integrative Omics, Guilin Medical University, Guilin, PR China
| | - Yu Liu
- School of Lingui Clinical Medicine, Guilin Medical University, Guilin, PR China; Laboratory of Environmental Pollution and Integrative Omics, Guilin Medical University, Guilin, PR China
| | - Wenjun Huang
- Laboratory of Environmental Pollution and Integrative Omics, Guilin Medical University, Guilin, PR China
| | - Rong Li
- Laboratory of Environmental Pollution and Integrative Omics, Guilin Medical University, Guilin, PR China.
| | - Keng Po Lai
- Laboratory of Environmental Pollution and Integrative Omics, Guilin Medical University, Guilin, PR China.
| |
Collapse
|
17
|
Ijomone OM, Iroegbu JD, Aschner M, Bornhorst J. Impact of environmental toxicants on p38- and ERK-MAPK signaling pathways in the central nervous system. Neurotoxicology 2021; 86:166-171. [PMID: 34389354 PMCID: PMC8440482 DOI: 10.1016/j.neuro.2021.08.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 08/06/2021] [Accepted: 08/07/2021] [Indexed: 12/16/2022]
Abstract
There are several candidate signalling pathways that mediate the response of the central nervous system (CNS) cells to environmental toxins. However, much is still to be learned on how these pathways modulate neurotoxicity. The mitogen-activated protein kinases (MAPKs) signalling pathways, which include the extracellular signal-regulated protein kinase (ERK) and the p38-MAPK, are potentially key pathways to regulate CNS responses to environmental toxins. The pathways play leading roles in the transmission of extracellular signals into the cell nucleus, leading to cell differentiation, cell growth, and apoptosis, to name a few. Moreover, exposure to environmental toxins induces p38- and ERK-MAPK activation, which leads to oxidative stress, inflammation, and apoptosis in the CNS. Here, we provide a concise review of the recent evidence demonstrating the role of p38- and ERK-MAPK signaling pathways and their downstream targets in the CNS following exposure to environmental toxicants such as metals, organophosphorus and persistent organic pollutants.
Collapse
Affiliation(s)
- Omamuyovwi M Ijomone
- Food Chemistry, Faculty of Mathematics and Natural Sciences, University of Wuppertal, Wuppertal, Germany; The Neuro- Lab, Department of Human Anatomy, School of Health and Health Technology, Federal University of Technology Akure, Akure, Nigeria.
| | - Joy D Iroegbu
- The Neuro- Lab, Department of Human Anatomy, School of Health and Health Technology, Federal University of Technology Akure, Akure, Nigeria
| | - Michael Aschner
- Departments of Molecular Pharmacology and Neurosciences, Albert Einstein College of Medicine, NY, USA
| | - Julia Bornhorst
- Food Chemistry, Faculty of Mathematics and Natural Sciences, University of Wuppertal, Wuppertal, Germany
| |
Collapse
|
18
|
Wang Z, Han S, Xu Z, Du P, Li X. Assessment on the adverse effects on different kinds of fish induced by methamphetamine during the natural attenuation process based on adverse outcome pathway. THE SCIENCE OF THE TOTAL ENVIRONMENT 2021; 780:146587. [PMID: 33773348 DOI: 10.1016/j.scitotenv.2021.146587] [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: 01/24/2021] [Revised: 03/13/2021] [Accepted: 03/15/2021] [Indexed: 06/12/2023]
Abstract
The adverse effects on model fish induced by methamphetamine (METH) have been revealed. However, the toxicity of METH on different kinds of non-model fish during the natural attenuation remained unclear. Hence, in this study, we for the first time established a static lab-scale aquatic ecosystem spiked with METH (initial levels at 25 μg/L) for 40 days to estimate its metabolism and toxicity in Chinese medaka, rosy bitterling, loach, and mosquito fish. The concentrations of METH in water and fish's brain were detected termly. The physiological functions, histopathology of brain, neurotransmitters contents, and expressions of associated genes of the four kinds of fish were determined at day 0, 20, and 40, respectively. The results indicated METH could be remarkably accumulated in fish brains with the distribution factor vs water (DFw) at 232.5-folds, and attenuated both in water and fish body during the exposure. METH caused physiological functions (i.e., swimming trajectories, locomotion distances, and feeding rates) disorders of the four kinds of fish, and stimulated surfacing behavior of loach. Tissue and macro/micromolecular biomarkers including histopathology, neurotransmitters (i.e., dopamine, serotonin, and norepinephrine), and mRNA, were similarly affected by METH. Mitogen-activated protein kinase (MAPKs) signaling pathway, P53-regulated apoptosis signaling pathway, N-methyl-d-aspartate-dopamine system, and mTOR signaling pathway of different kinds of fish were regulated by METH. Additionally, the impairments of the physiological and macromolecular indicators of fish could be alleviated as the natural attenuation of METH occurred. All the biomarkers, as well as the recovery effects during the exposure were integrated onto an adverse outcome pathway (AOP) framework. The key event was the micromolecular indicators (genes). The adverse outcomes at individual and population levels would result in the ecological consequences, implying the imperative to consider the natural attenuation process while assessing the environmental risk of METH.
Collapse
Affiliation(s)
- Zhenglu Wang
- College of Oceanography, Hohai University, Nanjing, Jiangsu 210098, PR China
| | - Sheng Han
- Laboratory for Earth Surface Processes, College of Urban and Environmental Sciences, Peking University, Beijing 100871, PR China
| | - Zeqiong Xu
- College of Geography and Environmental Sciences, Zhejiang Normal University, Jinhua, Zhejiang 321004, PR China
| | - Peng Du
- Beijing Key Laboratory of Urban Hydrological Cycle and Sponge City Technology, College of Water Sciences, Beijing Normal University, Beijing 100875, PR China.
| | - Xiqing Li
- Laboratory for Earth Surface Processes, College of Urban and Environmental Sciences, Peking University, Beijing 100871, PR China
| |
Collapse
|
19
|
Guo J, Wu P, Cao J, Luo Y, Chen J, Wang G, Guo W, Wang T, He X. The PFOS disturbed immunomodulatory functions via nuclear Factor-κB signaling in liver of zebrafish (Danio rerio). FISH & SHELLFISH IMMUNOLOGY 2019; 91:87-98. [PMID: 31082517 DOI: 10.1016/j.fsi.2019.05.018] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2018] [Revised: 05/08/2019] [Accepted: 05/09/2019] [Indexed: 05/18/2023]
Abstract
Excessive perfluorooctane sulfonate (PFOS) in natural water ecosystem has the potential to detrimentally affect immune system, but little is known of such effects or underlying mechanisms in fish. In the present study, we evaluated the effects of PFOS on growth performance, organizational microstructure, activities of immune-related enzymes and expressions of immune-related genes in male zebrafish (Danio rerio) exposed to different concentrations of 0, 0.02, 0.04 and 0.08 mg/L of PFOS for 7, 14, and 21 days or cotreatment with PFOS and PDTC to investigate the effects of PFOS on immune system and the potential toxic mechanisms caused by PFOS. The results indicated that PFOS accumulated in livers after exposure, and remarkably elevations were found in three exposure groups compared with the control group at three stages. The growth of the adult zebrafish in the experiments was significantly inhibited, the microstructures of liver were serious damaged. The ROS levels were remarkably increased. The activities of ACP, AKP, and lysozyme were obviously decreased, while the activities of MPO and NF-κB were significantly increased. The expressions of immune-related mRNA were significantly affected. After co-treatment with PFOS and PDTC, the growth inhibition, the morphological damage, the ROS induction, and the expressions of immune-related mRNA were reversed. Taken together, the results indicated that PFOS can significantly inhibit the growth, disturb the immune system by changing the normal structure of liver, the activities of immune-related enzymes, and a series of gene transcriptions involved in immune regulation in liver of male zebrafish. PFOS-induced pro-inflammatory effect of hepatocytes was observed, and the involvement of NF-κB signaling pathway was participated in its action mechanism. These findings provide further evidence that PFOS interferes with the immune regulation of liver of male zebrafish under in vivo conditions.
Collapse
Affiliation(s)
- Jinshu Guo
- State Key Laboratory of Ecological Animal Husbandry and Environment Veterinary Medicine, College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, 030801, shanxi, China
| | - Panhong Wu
- State Key Laboratory of Ecological Animal Husbandry and Environment Veterinary Medicine, College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, 030801, shanxi, China
| | - Jinling Cao
- State Key Laboratory of Ecological Animal Husbandry and Environment Veterinary Medicine, College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, 030801, shanxi, China.
| | - Yongju Luo
- Guangxi Key Laboratory of Aquatic Genetic Breeding and Healthy Aquaculture, Guangxi Acedemy of Fishery Science, Nanning, 530021, Guangxi, China.
| | - Jianjie Chen
- State Key Laboratory of Ecological Animal Husbandry and Environment Veterinary Medicine, College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, 030801, shanxi, China
| | - Guodong Wang
- State Key Laboratory of Ecological Animal Husbandry and Environment Veterinary Medicine, College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, 030801, shanxi, China
| | - Wenjing Guo
- State Key Laboratory of Ecological Animal Husbandry and Environment Veterinary Medicine, College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, 030801, shanxi, China
| | - Tianyu Wang
- State Key Laboratory of Ecological Animal Husbandry and Environment Veterinary Medicine, College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, 030801, shanxi, China
| | - Xinjing He
- State Key Laboratory of Ecological Animal Husbandry and Environment Veterinary Medicine, College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, 030801, shanxi, China
| |
Collapse
|
20
|
Zheng J, Fan R, Wu H, Yao H, Yan Y, Liu J, Ran L, Sun Z, Yi L, Dang L, Gan P, Zheng P, Yang T, Zhang Y, Tang T, Wang Y. Directed self-assembly of herbal small molecules into sustained release hydrogels for treating neural inflammation. Nat Commun 2019; 10:1604. [PMID: 30962431 PMCID: PMC6453967 DOI: 10.1038/s41467-019-09601-3] [Citation(s) in RCA: 214] [Impact Index Per Article: 35.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2018] [Accepted: 03/19/2019] [Indexed: 01/06/2023] Open
Abstract
Self-assembling natural drug hydrogels formed without structural modification and able to act as carriers are of interest for biomedical applications. A lack of knowledge about natural drug gels limits there current application. Here, we report on rhein, a herbal natural product, which is directly self-assembled into hydrogels through noncovalent interactions. This hydrogel shows excellent stability, sustained release and reversible stimuli-responses. The hydrogel consists of a three-dimensional nanofiber network that prevents premature degradation. Moreover, it easily enters cells and binds to toll-like receptor 4. This enables rhein hydrogels to significantly dephosphorylate IκBα, inhibiting the nuclear translocation of p65 at the NFκB signalling pathway in lipopolysaccharide-induced BV2 microglia. Subsequently, rhein hydrogels alleviate neuroinflammation with a long-lasting effect and little cytotoxicity compared to the equivalent free-drug in vitro. This study highlights a direct self-assembly hydrogel from natural small molecule as a promising neuroinflammatory therapy.
Collapse
Affiliation(s)
- Jun Zheng
- Key Laboratory of Hunan Province for Water Environment and Agriculture Product Safety, College of Chemistry and Chemical Engineering, Central South University, 410083, Changsha, China
| | - Rong Fan
- Institute of Integrative Medicine, Key Laboratory of Hunan Province for Liver Manifestation of Traditional Chinese Medicine, Xiangya Hospital, Central South University, 410008, Changsha, China
| | - Huiqiong Wu
- Key Laboratory of Hunan Province for Water Environment and Agriculture Product Safety, College of Chemistry and Chemical Engineering, Central South University, 410083, Changsha, China.,Key Laboratry of Materials Processing and Mold, Ministry of Education, Zhengzhou University, 450002, Zhengzhou, China
| | - Honghui Yao
- Xiangya School of Medicine, Central South University, 410013, Changsha, China
| | - Yujie Yan
- Xiangya School of Medicine, Central South University, 410013, Changsha, China
| | - Jiamiao Liu
- Xiangya School of Medicine, Central South University, 410013, Changsha, China
| | - Lu Ran
- Yunnan Food Safety Research Institute, Kunming University of Science and Technology, 650500, Kunming, China
| | - Zhifang Sun
- Key Laboratory of Hunan Province for Water Environment and Agriculture Product Safety, College of Chemistry and Chemical Engineering, Central South University, 410083, Changsha, China
| | - Lunzhao Yi
- Yunnan Food Safety Research Institute, Kunming University of Science and Technology, 650500, Kunming, China
| | - Li Dang
- Department of Chemistry and Key Laboratory for Preparation and Application of Ordered Structural Materials of Guangdong Province, Shantou University, 515063, Shantou, China
| | - Pingping Gan
- Department of Oncology, Xiangya Hospital, Central South University, 410008, Changsha, China
| | - Piao Zheng
- College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, 410208, Changsha, China
| | - Tilong Yang
- Southern University of Science and Technology, 518055, Shenzhen, China
| | - Yi Zhang
- Key Laboratory of Hunan Province for Water Environment and Agriculture Product Safety, College of Chemistry and Chemical Engineering, Central South University, 410083, Changsha, China. .,Institute of Integrative Medicine, Key Laboratory of Hunan Province for Liver Manifestation of Traditional Chinese Medicine, Xiangya Hospital, Central South University, 410008, Changsha, China.
| | - Tao Tang
- Institute of Integrative Medicine, Key Laboratory of Hunan Province for Liver Manifestation of Traditional Chinese Medicine, Xiangya Hospital, Central South University, 410008, Changsha, China.
| | - Yang Wang
- Institute of Integrative Medicine, Key Laboratory of Hunan Province for Liver Manifestation of Traditional Chinese Medicine, Xiangya Hospital, Central South University, 410008, Changsha, China.
| |
Collapse
|
21
|
Muripiti V, Brijesh L, Rachamalla HK, Marepally SK, Banerjee R, Patri SV. α-Tocopherol-ascorbic acid hybrid antioxidant based cationic amphiphile for gene delivery: Design, synthesis and transfection. Bioorg Chem 2018; 82:178-191. [PMID: 30326400 DOI: 10.1016/j.bioorg.2018.02.025] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 02/23/2018] [Accepted: 02/23/2018] [Indexed: 12/11/2022]
Abstract
Natural antioxidants and vitamins have potential to protect biological systems from peroxidative damage induced by peroxyl radicals, α-tocopherol (Vitamin E, lipid soluble) and ascorbic acid (vitamin C, water soluble), well known natural antioxidant molecules. In the present study we described the synthesis and biological evaluation of hybrid of these two natural antioxidants with each other via ammonium di-ethylether linker, Toc-As in gene delivery. Two control cationic lipids N14-As and Toc-NOH are designed in such a way that one is with ascorbic acid moiety and no tocopherol moiety; another is with tocopherol moiety and no ascorbic acid moiety respectively. All the three cationic lipids can form self-assembled aggregates. The antioxidant efficiencies of the three lipids were compared with free ascorbic acid. The cationic lipids (Toc-As, N14-As and Toc-NOH) were formulated individually with a well-known fusogenic co-lipid DOPE and characterization studies such as DNA binding, heparin displacement, size, charge, circular dichroism were performed. The biological characterization studies such as cell viability assay and in vitro transfection studies were carried out with the above formulations in HepG2, Neuro-2a, CHO andHEK-293T cell lines. The three formulations showed their transfection efficiencies with highest in Toc-As, moderate inN14-As and least in Toc-NOH. Interestingly, the transfection efficiency observed with the antioxidant based conjugated lipid Toc-As is found to be approximately two and half fold higher than the commercially available lipofectamine 2000 at 4:1 charge ratio in Hep G2 cell lines. In the other cell lines studied the efficiency of Toc-As is found to be either higher or similarly active compared to lipofectamine 2000. The physicochemical characterization results show that Toc-As lipid is showing maximum antioxidant potency, strong binding with pDNA, least size and optimal zeta potential. It is also found to be least toxic in all the cell lines studied especially in Neuro-2a cell lines when compared to other two lipids. In summary, the designed antioxidant lipid can be exploited as a delivering system for treating ROS related diseases such as malignancy, brain stroke, etc.
Collapse
Affiliation(s)
| | | | - Hari Krishnareddy Rachamalla
- Division of Lipid Science and Technology, Indian Institute of Chemical Technology, Hyderabad 500607, Telangana, India
| | | | - Rajkumar Banerjee
- Division of Lipid Science and Technology, Indian Institute of Chemical Technology, Hyderabad 500607, Telangana, India
| | | |
Collapse
|
22
|
Gan P, Zhang L, Chen Y, Zhang Y, Zhang F, Zhou X, Zhang X, Gao B, Zhen X, Zhang J, Zheng LT. Anti-inflammatory effects of glaucocalyxin B in microglia cells. J Pharmacol Sci 2015; 128:35-46. [PMID: 26003084 DOI: 10.1016/j.jphs.2015.04.005] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Revised: 04/07/2015] [Accepted: 04/09/2015] [Indexed: 11/29/2022] Open
Abstract
Over-activated microglia is involved in various kinds of neurodegenerative process including Parkinson, Alzheimer and HIV dementia. Suppression of microglial over activation has emerged as a novel strategy for treatment of neuroinflammation-based neurodegeneration. In the current study, anti-inflammatory and neuroprotective effects of the ent-kauranoid diterpenoids, which were isolated from the aerial parts of Rabdosia japonica (Burm. f.) var. glaucocalyx (Maxim.) Hara, were investigated in cultured microglia cells. Glaucocalyxin B (GLB), one of five ent-kauranoid diterpenoids, significantly decreased the generation of nitric oxide (NO), tumor necrosis factor (TNF)-α, interleukin (IL)-1β, cyclooxygenase (COX)-2 and inducible nitric oxide synthase (iNOS) in the lipopolysaccharide (LPS)-activated microglia cells. In addition, GLB inhibited activation of nuclear factor-κB (NF-κB), p38 mitogen-activated protein kinase (MAPK) and generation of reactive oxygen species (ROS) in LPS-activated microglia cells. Furthermore, GLB strongly induced the expression of heme oxygenase (HO)-1 in BV-2 microglia cells. Finally, GLB exhibited neuroprotective effect by preventing over-activated microglia induced neurotoxicity in a microglia/neuron co-culture model. Taken together, the present study demonstrated that the GLB possesses anti-nueroinflammatory activity, and might serve as a potential therapeutic agent for treating neuroinflammatory diseases.
Collapse
Affiliation(s)
- Ping Gan
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuropsychiatric Disorders & Department of Pharmacology, College of Pharmaceutical Sciences and the Collaborative Innovation Center for Brain Science, Soochow University, Suzhou 215123, PR China
| | - Li Zhang
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuropsychiatric Disorders & Department of Pharmacology, College of Pharmaceutical Sciences and the Collaborative Innovation Center for Brain Science, Soochow University, Suzhou 215123, PR China
| | - Yanke Chen
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuropsychiatric Disorders & Department of Pharmacology, College of Pharmaceutical Sciences and the Collaborative Innovation Center for Brain Science, Soochow University, Suzhou 215123, PR China
| | - Yu Zhang
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuropsychiatric Disorders & Department of Pharmacology, College of Pharmaceutical Sciences and the Collaborative Innovation Center for Brain Science, Soochow University, Suzhou 215123, PR China
| | - Fali Zhang
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuropsychiatric Disorders & Department of Pharmacology, College of Pharmaceutical Sciences and the Collaborative Innovation Center for Brain Science, Soochow University, Suzhou 215123, PR China
| | - Xiang Zhou
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuropsychiatric Disorders & Department of Pharmacology, College of Pharmaceutical Sciences and the Collaborative Innovation Center for Brain Science, Soochow University, Suzhou 215123, PR China
| | - Xiaohu Zhang
- Department of Medical Chemistry, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, PR China
| | - Bo Gao
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuropsychiatric Disorders & Department of Pharmacology, College of Pharmaceutical Sciences and the Collaborative Innovation Center for Brain Science, Soochow University, Suzhou 215123, PR China
| | - Xuechu Zhen
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuropsychiatric Disorders & Department of Pharmacology, College of Pharmaceutical Sciences and the Collaborative Innovation Center for Brain Science, Soochow University, Suzhou 215123, PR China
| | - Jian Zhang
- Department of Natural Medical Chemistry, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, PR China.
| | - Long Tai Zheng
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuropsychiatric Disorders & Department of Pharmacology, College of Pharmaceutical Sciences and the Collaborative Innovation Center for Brain Science, Soochow University, Suzhou 215123, PR China.
| |
Collapse
|