1
|
Xiao Y, Xiang W, Ma X, Zheng A, Rong D, Zhang N, Yang N, Bayram H, Lorimer GH, Wang J. Research Progress on the Correlation Between Atmospheric Particulate Matter and Autism. J Appl Toxicol 2024. [PMID: 39701085 DOI: 10.1002/jat.4722] [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: 06/29/2024] [Revised: 09/29/2024] [Accepted: 10/25/2024] [Indexed: 12/21/2024]
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder caused by the interaction of genetic and complex environmental factors. The prevalence of autism has dramatically increased in countries and regions undergoing rapid industrialization and urbanization. Recent studies have shown that particulate matter (PM) in air pollution affects the development of neurons and disrupts the function of the nervous system, leading to behavioral and cognitive problems and increasing the risk of ASD. However, research on the mechanism of environmental factors and ASD is still in its infancy. On this basis, we conducted a literature search and analysis to review epidemiological studies on the correlation between fine particulate matter (PM2.5) and inhalable particulate matter (PM10) and ASD. The signaling pathways and pathogenic mechanisms of PM in synaptic injury and neuroinflammation are presented, and the mechanism of the ASD candidate gene SHANK3 was reviewed. Additionally, the different sites of action of different particles in animal models and humans were highlighted, and the differences of their effects on the pathogenesis of ASD were explained. We summarized the aetiology and mechanisms of PM-induced autism and look forward to future research breakthroughs in improved assessment methods, multidisciplinary alliances and high-tech innovations.
Collapse
Affiliation(s)
- Yaqian Xiao
- Cooperative Innovation Center of Industrial Fermentation, Ministry of Education & Hubei Province, Hubei University of Technology, Wuhan, Hubei, China
- International Center for Redox Biology & Precision Medicine of Hubei Province, Hubei University of Technology, Wuhan, Hubei, China
- Autism & Depression Diagnosis and Intervention Institute, School of Biological Engineering and Food, Hubei University of Technology, Wuhan, Hubei, China
| | - Wang Xiang
- Cooperative Innovation Center of Industrial Fermentation, Ministry of Education & Hubei Province, Hubei University of Technology, Wuhan, Hubei, China
- International Center for Redox Biology & Precision Medicine of Hubei Province, Hubei University of Technology, Wuhan, Hubei, China
- Autism & Depression Diagnosis and Intervention Institute, School of Biological Engineering and Food, Hubei University of Technology, Wuhan, Hubei, China
| | - Xuerui Ma
- Cooperative Innovation Center of Industrial Fermentation, Ministry of Education & Hubei Province, Hubei University of Technology, Wuhan, Hubei, China
- International Center for Redox Biology & Precision Medicine of Hubei Province, Hubei University of Technology, Wuhan, Hubei, China
- Autism & Depression Diagnosis and Intervention Institute, School of Biological Engineering and Food, Hubei University of Technology, Wuhan, Hubei, China
| | - Aijia Zheng
- Cooperative Innovation Center of Industrial Fermentation, Ministry of Education & Hubei Province, Hubei University of Technology, Wuhan, Hubei, China
- International Center for Redox Biology & Precision Medicine of Hubei Province, Hubei University of Technology, Wuhan, Hubei, China
- Autism & Depression Diagnosis and Intervention Institute, School of Biological Engineering and Food, Hubei University of Technology, Wuhan, Hubei, China
| | - Dechang Rong
- Cooperative Innovation Center of Industrial Fermentation, Ministry of Education & Hubei Province, Hubei University of Technology, Wuhan, Hubei, China
- International Center for Redox Biology & Precision Medicine of Hubei Province, Hubei University of Technology, Wuhan, Hubei, China
- Autism & Depression Diagnosis and Intervention Institute, School of Biological Engineering and Food, Hubei University of Technology, Wuhan, Hubei, China
| | - Nimeng Zhang
- Cooperative Innovation Center of Industrial Fermentation, Ministry of Education & Hubei Province, Hubei University of Technology, Wuhan, Hubei, China
- International Center for Redox Biology & Precision Medicine of Hubei Province, Hubei University of Technology, Wuhan, Hubei, China
- Autism & Depression Diagnosis and Intervention Institute, School of Biological Engineering and Food, Hubei University of Technology, Wuhan, Hubei, China
| | - Ning Yang
- Cooperative Innovation Center of Industrial Fermentation, Ministry of Education & Hubei Province, Hubei University of Technology, Wuhan, Hubei, China
- International Center for Redox Biology & Precision Medicine of Hubei Province, Hubei University of Technology, Wuhan, Hubei, China
- Autism & Depression Diagnosis and Intervention Institute, School of Biological Engineering and Food, Hubei University of Technology, Wuhan, Hubei, China
| | - Hasan Bayram
- Department of Pulmonary Medicine, School of Medicine, Koc University, Istanbul, Turkey
| | - George H Lorimer
- Department of Chemistry, University of Maryland, College Park, Maryland, USA
| | - Jun Wang
- Cooperative Innovation Center of Industrial Fermentation, Ministry of Education & Hubei Province, Hubei University of Technology, Wuhan, Hubei, China
- International Center for Redox Biology & Precision Medicine of Hubei Province, Hubei University of Technology, Wuhan, Hubei, China
- Autism & Depression Diagnosis and Intervention Institute, School of Biological Engineering and Food, Hubei University of Technology, Wuhan, Hubei, China
| |
Collapse
|
2
|
Bove M, Sikora V, Santoro M, Agosti LP, Palmieri MA, Dimonte S, Tucci P, Schiavone S, Morgese MG, Trabace L. Sex differences in the BTBR idiopathic mouse model of autism spectrum disorders: Behavioural and redox-related hippocampal alterations. Neuropharmacology 2024; 260:110134. [PMID: 39208979 DOI: 10.1016/j.neuropharm.2024.110134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 08/24/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
Autism spectrum disorders (ASD) are highly heterogeneous neurodevelopmental diseases. Epidemiological data report that males have been diagnosed with autism more frequently than females. However, recent studies hypothesize that females' low incidence might be underestimated due to standard clinical measures of ASD behavioural symptoms, mostly derived from males. Indeed, up to now, ASD mouse models focused mainly on males, considering the prevalence of the diagnosis in that sex. Regarding ASD aetiopathogenesis, it has been recently reported that oxidative stress might be implicated in its onset and development, suggesting an association with ASD typical repetitive behaviours that still need to be disentangled. Here, we investigated possible behavioural and molecular sex-related differences by using the BTBR mouse model of idiopathic ASD. To this aim, animals were exposed to behavioural tests related to different ASD core symptoms and comorbidities, i.e. stereotyped repertoire, social dysfunctions, hyperlocomotion and risk-taking behaviours. Moreover, we analyzed hippocampal levels of pro-oxidant and anti-oxidant enzymes, together with biomarkers of oxidative stress and lipid peroxidation. Our results showed that BTBR females did not display the same patterns for repetitive behaviours as the male counterpart. From a biomolecular point of view, we found an increase in oxidative stress and pro-oxidant enzymes, accompanied by deficient enzymatic anti-oxidant response, only in BTBR males compared to C57BL/6 male mice, while no differences were retrieved in females. Overall, our study suggests that in females there is an urgent need to depict the distinct ASD symptomatology, accompanied by the identification of sex-specific pharmacological targets.
Collapse
Affiliation(s)
- Maria Bove
- Department of Clinical and Experimental Medicine, University of Foggia, Via Napoli 20, 71122, Foggia, Italy
| | - Vladyslav Sikora
- Department of Clinical and Experimental Medicine, University of Foggia, Via Napoli 20, 71122, Foggia, Italy; Department of Pathology, Sumy State University, 40007, Sumy, Ukraine
| | - Martina Santoro
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, 00185, Rome, Italy
| | - Lisa Pia Agosti
- Department of Clinical and Experimental Medicine, University of Foggia, Via Napoli 20, 71122, Foggia, Italy
| | - Maria Adelaide Palmieri
- Department of Clinical and Experimental Medicine, University of Foggia, Via Napoli 20, 71122, Foggia, Italy
| | - Stefania Dimonte
- Department of Clinical and Experimental Medicine, University of Foggia, Via Napoli 20, 71122, Foggia, Italy
| | - Paolo Tucci
- Department of Clinical and Experimental Medicine, University of Foggia, Via Napoli 20, 71122, Foggia, Italy
| | - Stefania Schiavone
- Department of Clinical and Experimental Medicine, University of Foggia, Via Napoli 20, 71122, Foggia, Italy
| | - Maria Grazia Morgese
- Department of Clinical and Experimental Medicine, University of Foggia, Via Napoli 20, 71122, Foggia, Italy
| | - Luigia Trabace
- Department of Clinical and Experimental Medicine, University of Foggia, Via Napoli 20, 71122, Foggia, Italy.
| |
Collapse
|
3
|
Janner DE, Poetini MR, Musachio EAS, Chaves NSG, Meichtry LB, Fernandes EJ, Mustafa MMD, De Carvalho AS, Gonçalves OH, Leimann FV, de Freitas RA, Prigol M, Guerra GP. Neurodevelopmental changes in Drosophila melanogaster are restored by treatment with lutein-loaded nanoparticles: Positive modulation of neurochemical and behavioral parameters. Comp Biochem Physiol C Toxicol Pharmacol 2024; 285:109998. [PMID: 39106915 DOI: 10.1016/j.cbpc.2024.109998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 07/31/2024] [Accepted: 08/01/2024] [Indexed: 08/09/2024]
Abstract
Neurodevelopmental disorders, such as autism spectrum disorder (ASD) and attention deficit hyperactivity disorder (ADHD), are characterized by persistent changes in communication and social interaction, as well as restricted and stereotyped patterns of behavior. The complex etiology of these disorders possibly combines the effects of multiple genes and environmental factors. Hence, exposure to insecticides such as imidacloprid (IMI) has been used to replicate the changes observed in these disorders. Lutein is known for its anti-inflammatory and antioxidant properties and is associated with neuroprotective effects. Therefore, the aim of this study was to evaluate the protective effect of lutein-loaded nanoparticles, along with their mechanisms of action, on Drosophila melanogaster offspring exposed to IMI-induced damage. To simulate the neurodevelopmental disorder model, flies were exposed to a diet containing IMI for 7 days. Posteriorly, their offspring were exposed to a diet containing lutein-loaded nanoparticles for a period of 24 h, and male and female flies were subjected to behavioral and biochemical evaluations. Treatment with lutein-loaded nanoparticles reversed the parameters of hyperactivity, aggressiveness, social interaction, repetitive movements, and anxiety in the offspring of flies exposed to IMI. It also protected markers of oxidative stress and cell viability, in addition to preventing the reduction of Nrf2 and Shank3 immunoreactivity. These results demonstrate that the damage induced by exposure to IMI was restored through treatment with lutein-loaded nanoparticles, elucidating lutein's mechanisms of action as a therapeutic agent, which, after further studies, can become a co-adjuvant in the treatment of neurodevelopmental disorders, such as ASD and ADHD.
Collapse
Affiliation(s)
- Dieniffer Espinosa Janner
- Laboratory of Pharmacological and Toxicological Evaluations Applied to Bioactive Molecules - LaftamBio, Federal University of Pampa - Campus Itaqui, 97650-000 Itaqui, RS, Brazil; Postgraduate Program in Biochemistry, Federal University of Pampa - Campus Uruguaiana, 97508-000 Uruguaiana, RS, Brazil
| | - Márcia Rósula Poetini
- Laboratory of Pharmacological and Toxicological Evaluations Applied to Bioactive Molecules - LaftamBio, Federal University of Pampa - Campus Itaqui, 97650-000 Itaqui, RS, Brazil; Postgraduate Program in Biochemistry, Federal University of Pampa - Campus Uruguaiana, 97508-000 Uruguaiana, RS, Brazil
| | - Elize Aparecida Santos Musachio
- Laboratory of Pharmacological and Toxicological Evaluations Applied to Bioactive Molecules - LaftamBio, Federal University of Pampa - Campus Itaqui, 97650-000 Itaqui, RS, Brazil; Postgraduate Program in Biochemistry, Federal University of Pampa - Campus Uruguaiana, 97508-000 Uruguaiana, RS, Brazil
| | - Nathalie Savedra Gomes Chaves
- Laboratory of Pharmacological and Toxicological Evaluations Applied to Bioactive Molecules - LaftamBio, Federal University of Pampa - Campus Itaqui, 97650-000 Itaqui, RS, Brazil; Postgraduate Program in Biochemistry, Federal University of Pampa - Campus Uruguaiana, 97508-000 Uruguaiana, RS, Brazil
| | - Luana Barreto Meichtry
- Laboratory of Pharmacological and Toxicological Evaluations Applied to Bioactive Molecules - LaftamBio, Federal University of Pampa - Campus Itaqui, 97650-000 Itaqui, RS, Brazil; Postgraduate Program in Biochemistry, Federal University of Pampa - Campus Uruguaiana, 97508-000 Uruguaiana, RS, Brazil
| | - Eliana Jardim Fernandes
- Laboratory of Pharmacological and Toxicological Evaluations Applied to Bioactive Molecules - LaftamBio, Federal University of Pampa - Campus Itaqui, 97650-000 Itaqui, RS, Brazil; Postgraduate Program in Biochemistry, Federal University of Pampa - Campus Uruguaiana, 97508-000 Uruguaiana, RS, Brazil
| | - Mustafa Munir Dahleh Mustafa
- Laboratory of Pharmacological and Toxicological Evaluations Applied to Bioactive Molecules - LaftamBio, Federal University of Pampa - Campus Itaqui, 97650-000 Itaqui, RS, Brazil; Postgraduate Program in Biochemistry, Federal University of Pampa - Campus Uruguaiana, 97508-000 Uruguaiana, RS, Brazil
| | - Amarilis Santos De Carvalho
- Graduate Program in Food Technology, Federal Technological University of Paraná - Campus Campo Mourão, 87301-006 Campo Mourão, PR, Brazil
| | - Odinei Hess Gonçalves
- Graduate Program in Food Technology, Federal Technological University of Paraná - Campus Campo Mourão, 87301-006 Campo Mourão, PR, Brazil
| | - Fernanda Vitória Leimann
- Graduate Program in Food Technology, Federal Technological University of Paraná - Campus Campo Mourão, 87301-006 Campo Mourão, PR, Brazil
| | | | - Marina Prigol
- Laboratory of Pharmacological and Toxicological Evaluations Applied to Bioactive Molecules - LaftamBio, Federal University of Pampa - Campus Itaqui, 97650-000 Itaqui, RS, Brazil; Postgraduate Program in Biochemistry, Federal University of Pampa - Campus Uruguaiana, 97508-000 Uruguaiana, RS, Brazil
| | - Gustavo Petri Guerra
- Laboratory of Pharmacological and Toxicological Evaluations Applied to Bioactive Molecules - LaftamBio, Federal University of Pampa - Campus Itaqui, 97650-000 Itaqui, RS, Brazil; Postgraduate Program in Biochemistry, Federal University of Pampa - Campus Uruguaiana, 97508-000 Uruguaiana, RS, Brazil.
| |
Collapse
|
4
|
Acton S, O'Donnell MM, Periyasamy K, Dixit B, Eishingdrelo H, Hill C, Paul Ross R, Chesnel L. LPA3 agonist-producing Bacillus velezensis ADS024 is efficacious in multiple neuroinflammatory disease models. Brain Behav Immun 2024; 121:384-402. [PMID: 39147172 DOI: 10.1016/j.bbi.2024.08.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 08/08/2024] [Accepted: 08/10/2024] [Indexed: 08/17/2024] Open
Abstract
Neuroinflammation is a common component of neurological disorders. In the gut-brain-immune axis, bacteria and their metabolites are now thought to play a role in the modulation of the nervous and immune systems which may impact neuroinflammation. In this respect, commensal bacteria of humans have recently been shown to produce metabolites that mimic endogenous G-protein coupled receptor (GPCR) ligands. To date, it has not been established whether plant commensal bacteria, which may be ingested by animals including humans, can impact the gut-brain-immune axis via GPCR agonism. We screened an isopropanol (IPA) extract of the plant commensal Bacillus velezensis ADS024, a non-engrafting live biotherapeutic product (LBP) with anti-inflammatory properties isolated from human feces, against a panel of 168 GPCRs and identified strong agonism of the lysophosphatidic acid (LPA) receptor LPA3. The ADS024 IPA extracted material (ADS024-IPA) did not agonize LPA2, and only very weakly agonized LPA1. The agonism of LPA3 was inhibited by the reversible LPA1/3 antagonist Ki16425. ADS024-IPA signaled downstream of LPA3 through G-protein-induced calcium release, recruitment of β-arrestin, and recruitment of the neurodegeneration-associated proteins 14-3-3γ, ε and ζ but did not recruit the β isoform. Since LPA3 agonism was previously indirectly implicated in the reduction of pathology in models of Parkinson's disease (PD) and multiple sclerosis (MS) by use of the nonselective antagonist Ki16425, and since we identified an LPA3-specific agonist within ADS024, we sought to examine whether LPA3 might indeed be part of a broad underlying mechanism to control neuroinflammation. We tested oral treatment of ADS024 in multiple models of neuroinflammatory diseases using three models of PD, two models of MS, and a model each of amyotrophic lateral sclerosis (ALS), Huntington's disease (HD), and chemo-induced peripheral neuropathy (CIPN). ADS024 treatment improved model-specific functional effects including improvements in motor movement, breathing and swallowing, and allodynia suggesting that ADS024 treatment impacted a universal underlying neuroinflammatory mechanism regardless of the initiating cause of disease. We used the MOG-EAE mouse model to examine early events after disease initiation and found that ADS024 attenuated the increase in circulating lymphocytes and changes in neutrophil subtypes, and ADS024 attenuated the early loss of cell-surface LPA3 receptor expression on circulating white blood cells. ADS024 efficacy was partially inhibited by Ki16425 in vivo suggesting LPA3 may be part of its mechanism. Altogether, these data suggest that ADS024 and its LPA3 agonism activity should be investigated further as a possible treatment for diseases with a neuroinflammatory component.
Collapse
Affiliation(s)
| | | | | | | | | | - Colin Hill
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - R Paul Ross
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | | |
Collapse
|
5
|
Hirai T, Umeda N, Harada T, Okumura A, Nakayasu C, Ohto‐Nakanishi T, Tsuchiya KJ, Nishimura T, Matsuzaki H. Arachidonic acid-derived dihydroxy fatty acids in neonatal cord blood relate symptoms of autism spectrum disorders and social adaptive functioning: Hamamatsu Birth Cohort for Mothers and Children (HBC Study). Psychiatry Clin Neurosci 2024; 78:546-557. [PMID: 39041066 PMCID: PMC11488600 DOI: 10.1111/pcn.13710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 05/21/2024] [Accepted: 06/16/2024] [Indexed: 07/24/2024]
Abstract
AIM Autism spectrum disorder (ASD) is associated with abnormal lipid metabolism, such as a high total ratio of omega-6 to omega-3 in polyunsaturated fatty acids (PUFAs). PUFAs are metabolized to epoxy fatty acids by cytochrome P450 (CYP); then, dihydroxy fatty acid is produced by soluble epoxide hydrolase. This study examined the association between PUFA metabolites in the cord blood and ASD symptoms and adaptive functioning in children. METHODS This prospective cohort study utilized cord blood to quantify PUFA metabolites of the CYP pathway. The Autism Diagnostic Observation Schedule (ADOS-2) and Vineland Adaptive Behaviors Scales, Second Edition (VABS-II) were used to assess subsequent ASD symptoms and adaptive functioning in children at 6 years. The analysis included 200 children and their mothers. RESULTS Arachidonic acid-derived diols, 11,12-diHETrE was found to impact ASD symptom severity on the ADOS-2-calibrated severity scores and impairment in the socialization domain as assessed by the VABS-II (P = 0.0003; P = 0.004, respectively). High levels of 11,12-diHETrE impact social affect in ASD symptoms (P = 0.002), while low levels of 8,9-diHETrE impact repetitive/restrictive behavior (P = 0.003). Notably, there was specificity in the association between diHETrE and ASD symptoms, especially in girls. CONCLUSION These findings suggest that the dynamics of diHETrE during the fetal period is important in the developmental trajectory of children after birth. Given that the role of diol metabolites in neurodevelopment in vivo is completely uncharacterized, the results of this study provide important insight into the role of diHETrE and ASD pathophysiology.
Collapse
Affiliation(s)
- Takaharu Hirai
- Department of Psychiatric and Mental Health Nursing, School of NursingUniversity of FukuiEiheijiJapan
- Life Science Innovation CenterUniversity of FukuiEiheijiJapan
| | - Naoko Umeda
- Life Science Innovation CenterUniversity of FukuiEiheijiJapan
- Department of Maternal and Child Health Nursing, School of NursingUniversity of FukuiEiheijiJapan
| | - Taeko Harada
- Research Center for Child Mental DevelopmentHamamatsu University School of MedicineHamamatsuJapan
- United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of MedicineChiba University and University of FukuiSuitaJapan
| | - Akemi Okumura
- Research Center for Child Mental DevelopmentHamamatsu University School of MedicineHamamatsuJapan
- United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of MedicineChiba University and University of FukuiSuitaJapan
| | - Chikako Nakayasu
- Research Center for Child Mental DevelopmentHamamatsu University School of MedicineHamamatsuJapan
| | | | - Kenji J. Tsuchiya
- Research Center for Child Mental DevelopmentHamamatsu University School of MedicineHamamatsuJapan
- United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of MedicineChiba University and University of FukuiSuitaJapan
| | - Tomoko Nishimura
- Research Center for Child Mental DevelopmentHamamatsu University School of MedicineHamamatsuJapan
- United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of MedicineChiba University and University of FukuiSuitaJapan
| | - Hideo Matsuzaki
- United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of MedicineChiba University and University of FukuiSuitaJapan
- Research Center for Child Mental DevelopmentUniversity of FukuiEiheijiJapan
| |
Collapse
|
6
|
Seneff S, Kyriakopoulos AM, Nigh G. Is autism a PIN1 deficiency syndrome? A proposed etiological role for glyphosate. J Neurochem 2024; 168:2124-2146. [PMID: 38808598 DOI: 10.1111/jnc.16140] [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: 03/30/2024] [Revised: 05/13/2024] [Accepted: 05/14/2024] [Indexed: 05/30/2024]
Abstract
Autism is a neurodevelopmental disorder, the prevalence of which has increased dramatically in the United States over the past two decades. It is characterized by stereotyped behaviors and impairments in social interaction and communication. In this paper, we present evidence that autism can be viewed as a PIN1 deficiency syndrome. Peptidyl-prolyl cis/trans isomerase, NIMA-Interacting 1 (PIN1) is a peptidyl-prolyl cis/trans isomerase, and it has widespread influences in biological organisms. Broadly speaking, PIN1 deficiency is linked to many neurodegenerative diseases, whereas PIN1 over-expression is linked to cancer. Death-associated protein kinase 1 (DAPK1) strongly inhibits PIN1, and the hormone melatonin inhibits DAPK1. Melatonin deficiency is strongly linked to autism. It has recently been shown that glyphosate exposure to rats inhibits melatonin synthesis as a result of increased glutamate release from glial cells and increased expression of metabotropic glutamate receptors. Glyphosate's inhibition of melatonin leads to a reduction in PIN1 availability in neurons. In this paper, we show that PIN1 deficiency can explain many of the unique morphological features of autism, including increased dendritic spine density, missing or thin corpus callosum, and reduced bone density. We show how PIN1 deficiency disrupts the functioning of powerful high-level signaling molecules, such as nuclear factor erythroid 2-related factor 2 (NRF2) and p53. Dysregulation of both of these proteins has been linked to autism. Severe depletion of glutathione in the brain resulting from chronic exposure to oxidative stressors and extracellular glutamate leads to oxidation of the cysteine residue in PIN1, inactivating the protein and further contributing to PIN1 deficiency. Impaired autophagy leads to increased sensitivity of neurons to ferroptosis. It is imperative that further research be conducted to experimentally validate whether the mechanisms described here take place in response to chronic glyphosate exposure and whether this ultimately leads to autism.
Collapse
Affiliation(s)
- Stephanie Seneff
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | | | - Greg Nigh
- Immersion Health, Portland, Oregon, USA
| |
Collapse
|
7
|
Alshehri S, Ahmad SF, Albekairi NA, Alqarni SS, Al-Harbi NO, Al-Ayadhi LY, Attia SM, Alfardan AS, Bakheet SA, Nadeem A. Thioredoxin 1 and Thioredoxin Reductase 1 Redox System Is Dysregulated in Neutrophils of Subjects with Autism: In Vitro Effects of Environmental Toxicant, Methylmercury. TOXICS 2023; 11:739. [PMID: 37755749 PMCID: PMC10536321 DOI: 10.3390/toxics11090739] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 08/25/2023] [Accepted: 08/25/2023] [Indexed: 09/28/2023]
Abstract
Autism spectrum disorder (ASD) is a complex developmental disorder in children that results in abnormal communicative and verbal behaviors. Exposure to heavy metals plays a significant role in the pathogenesis or progression of ASD. Mercury compounds pose significant risk for the development of ASD as children are more exposed to environmental toxicants. Increased concentration of mercury compounds has been detected in different body fluids/tissues in ASD children, which suggests an association between mercury exposure and ASD. Thioredoxin1 (Trx1) and thioredoxin reductase1 (TrxR1) redox system plays a crucial role in detoxification of oxidants generated in different immune cells. However, the effect of methylmercury and the Nrf2 activator sulforaphane on the Trx1/TrxR1 antioxidant system in neutrophils of ASD subjects has not been studied previously. Therefore, this study examined the effect of methylmercury on Trx1/TrxR1 expression, TrxR activity, nitrotyrosine, and ROS in neutrophils of ASD and TDC subjects. Our study shows that Trx1/TrxR1 protein expression is dysregulated in ASD subjects as compared to the TDC group. Further, methylmercury treatment significantly inhibits the activity of TrxR in both ASD and TDC groups. Inhibition of TrxR by mercury is associated with upregulation of the Trx1 protein in TDC neutrophils but not in ASD neutrophils. Furthermore, ASD neutrophils have exaggerated ROS production after exposure to methylmercury, which is much greater in magnitude than TDC neutrophils. Sulforaphane reversed methylmercury-induced effects on neutrophils through Nrf2-mediated induction of the Trx1/TrxR1 system. These observations suggest that exposure to the environmental toxicant methylmercury may elevate systemic oxidative inflammation due to a dysregulated Trx1/TrxR1 redox system in the neutrophils of ASD subjects, which may play a role in the progression of ASD.
Collapse
Affiliation(s)
- Samiyah Alshehri
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Sheikh F. Ahmad
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Norah A. Albekairi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Sana S. Alqarni
- Department of Medical Laboratory Science, College of Applied Medical Sciences, King Saud University, Riyadh 11451, Saudi Arabia
| | - Naif O. Al-Harbi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Laila Y. Al-Ayadhi
- Department of Physiology, College of Medicine, King Saud University, Riyadh 11451, Saudi Arabia
| | - Sabry M. Attia
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Ali S. Alfardan
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Saleh A. Bakheet
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Ahmed Nadeem
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| |
Collapse
|
8
|
Buchanan E, Mahony C, Bam S, Jaffer M, Macleod S, Mangali A, van der Watt M, de Wet S, Theart R, Jacobs C, Loos B, O'Ryan C. Propionic acid induces alterations in mitochondrial morphology and dynamics in SH-SY5Y cells. Sci Rep 2023; 13:13248. [PMID: 37582965 PMCID: PMC10427685 DOI: 10.1038/s41598-023-40130-8] [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: 12/30/2022] [Accepted: 08/04/2023] [Indexed: 08/17/2023] Open
Abstract
Propionic acid (PPA) is used to study the role of mitochondrial dysfunction in neurodevelopmental conditions like autism spectrum disorders. PPA is known to disrupt mitochondrial biogenesis, metabolism, and turnover. However, the effect of PPA on mitochondrial dynamics, fission, and fusion remains challenging to study due to the complex temporal nature of these mechanisms. Here, we use complementary quantitative visualization techniques to examine how PPA influences mitochondrial ultrastructure, morphology, and dynamics in neuronal-like SH-SY5Y cells. PPA (5 mM) induced a significant decrease in mitochondrial area (p < 0.01), Feret's diameter and perimeter (p < 0.05), and in area2 (p < 0.01). Mitochondrial event localiser analysis demonstrated a significant increase in fission and fusion events (p < 0.05) that preserved mitochondrial network integrity under stress. Moreover, mRNA expression of cMYC (p < 0.0001), NRF1 (p < 0.01), TFAM (p < 0.05), STOML2 (p < 0.0001), and OPA1 (p < 0.01) was significantly decreased. This illustrates a remodeling of mitochondrial morphology, biogenesis, and dynamics to preserve function under stress. Our data provide new insights into the influence of PPA on mitochondrial dynamics and highlight the utility of visualization techniques to study the complex regulatory mechanisms involved in the mitochondrial stress response.
Collapse
Affiliation(s)
- Erin Buchanan
- Department of Molecular and Cell Biology, University of Cape Town, Cape Town, 7700, South Africa
| | - Caitlyn Mahony
- Department of Molecular and Cell Biology, University of Cape Town, Cape Town, 7700, South Africa
| | - Sophia Bam
- Department of Molecular and Cell Biology, University of Cape Town, Cape Town, 7700, South Africa
| | - Mohamed Jaffer
- Electron Microscope Unit, University of Cape Town, Cape Town, 7700, South Africa
| | - Sarah Macleod
- Department of Molecular and Cell Biology, University of Cape Town, Cape Town, 7700, South Africa
| | - Asandile Mangali
- Department of Physiological Sciences, Stellenbosch University, Matieland, Stellenbosch, 7602, South Africa
| | - Mignon van der Watt
- Department of Molecular and Cell Biology, University of Cape Town, Cape Town, 7700, South Africa
| | - Sholto de Wet
- Department of Physiological Sciences, Stellenbosch University, Matieland, Stellenbosch, 7602, South Africa
| | - Rensu Theart
- Department of Electrical and Electronic Engineering, Stellenbosch University, Matieland, Stellenbosch, 7602, South Africa
| | - Caron Jacobs
- Department of Pathology, Wellcome Centre for Infectious Diseases Research in Africa and IDM Microscopy Platform, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, 7700, South Africa
| | - Ben Loos
- Department of Physiological Sciences, Stellenbosch University, Matieland, Stellenbosch, 7602, South Africa
| | - Colleen O'Ryan
- Department of Molecular and Cell Biology, University of Cape Town, Cape Town, 7700, South Africa.
- Neuroscience Institute, University of Cape Town, Cape Town, 7700, South Africa.
| |
Collapse
|
9
|
Algahtani MM, Ahmad SF, Alkharashi LA, Al-Harbi NO, Alanazi WA, Alhamed AS, Attia SM, Bakheet SA, Ibrahim KE, Nadeem A. Exposure to Methylmercury at Juvenile Stage Worsens Autism-like Symptoms in Adult BTBR T+tf/J Mice Due to Lack of Nuclear Factor Erythroid 2-Related Factor 2 Signaling Upregulation in Periphery and Brain. TOXICS 2023; 11:546. [PMID: 37368646 DOI: 10.3390/toxics11060546] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 06/16/2023] [Accepted: 06/19/2023] [Indexed: 06/29/2023]
Abstract
Autism spectrum disorder (ASD) is a multifaceted developmental condition that first appears in infancy. The condition is characterized by recurrent patterns in behavior and impairments in social and vocalization abilities. Methylmercury is a toxic environmental pollutant, and its derivatives are the major source of organic mercury to human beings. Inorganic mercury, which is released from a variety of pollutants into oceans, rivers, and streams, is transformed into methylmercury by bacteria and plankton in the water, which later builds up in fish and shellfish, and then enters humans through the consumption of fish and shellfish and increases the risk of developing ASD by disturbing the oxidant-antioxidant balance. However, there has been no prior research to determine the effect of juvenile exposure of methylmercury chloride on adult BTBR mice. Therefore, the current study evaluated the effect of methylmercury chloride administered during the juvenile stage on autism-like behavior (three-chambered sociability, marble burying, self-grooming tests) and oxidant-antioxidant balance (specifically Nrf2, HO-1, SOD-1, NF-kB, iNOS, MPO, and 3-nitrotyrosine) in the peripheral neutrophils and cortex of adult BTBR and C57BL/6 (B6) mice. Our results show that exposure to methylmercury chloride at a juvenile stage results in autism-like symptoms in adult BTBR mice which are related to a lack of upregulation of the Nrf2 signaling pathway as demonstrated by no significant changes in the expression of Nrf2, HO-1, and SOD-1 in the periphery and cortex. On the other hand, methylmercury chloride administration at a juvenile stage increased oxidative inflammation as depicted by a significant increase in the levels of NF-kB, iNOS, MPO, and 3-nitrotyrosine in the periphery and cortex of adult BTBR mice. This study suggests that juvenile exposure to methylmercury chloride contributes to the worsening of autism-like behavior in adult BTBR mice through the disruption of the oxidant-antioxidant balance in the peripheral compartment and CNS. Strategies that elevate Nrf2 signaling may be useful to counteract toxicant-mediated worsening of ASD and may improve quality of life.
Collapse
Affiliation(s)
- Mohammad M Algahtani
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Sheikh F Ahmad
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Layla A Alkharashi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Naif O Al-Harbi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Wael A Alanazi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Abdullah S Alhamed
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Sabry M Attia
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Saleh A Bakheet
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Khalid E Ibrahim
- Department of Zoology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Ahmed Nadeem
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| |
Collapse
|
10
|
Sun X, Liu N, Sun C, Xu Y, Ding D, Kong J. The inhibitory effect of vitamin D on myocardial homocysteine levels involves activation of Nrf2-mediated methionine synthase. J Steroid Biochem Mol Biol 2023; 231:106303. [PMID: 36990164 DOI: 10.1016/j.jsbmb.2023.106303] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 02/18/2023] [Accepted: 03/26/2023] [Indexed: 03/30/2023]
Abstract
BACKGROUND Homocysteine (Hcy) is a synthetic amino acid containing sulfhydryl group, which is an intermediate product of the deep metabolic pathway of methionine and cysteine. The abnormal increase in fasting plasma total Hcy concentration caused by various factors is called hyperhomocysteine (HHcy). HHcy is closely relevant to the occurrence and progression of diverse cardiovascular and cerebrovascular diseases, such as coronary heart disease, hypertension and diabetes, etc. Vitamin D/vitamin D receptor (VDR) pathway is pointed out that prevent cardiovascular disease by reducing serum homocysteine levels. Our research is designed to explore the potential mechanism of vitamin D in the prevention and treatment of HHcy. METHODS AND RESULTS The Hcy and 25(OH)D3 levels in mouse myocardial tissue, serum or myocardial cells were detected using ELISA kits. The expression levels of VDR, Nrf2 and methionine synthase (MTR) were observed using Western blotting, immunohistochemistry and real time polymerase chain reaction (PCR). General information of the mice, including diet, water intake and body weight, was recorded. Vitamin D up-regulated the mRNA and protein expression of Nrf2 and MTR in mouse myocardial tissue and cells. CHIP assay determined that the combination of Nrf2 binding to the S1 site of the MTR promoter in cardiomyocytes using traditional PCR and real time PCR. Dual Luciferase Assay was applied to detect the transcriptional control of Nrf2 on MTR. The up-regulation effect of Nrf2 on MTR was verified by Nrf2 knockout and overexpression in cardiomyocytes. The role of Nrf2 in vitamin D inhibition of Hcy was revealed using Nrf2-knockdown HL-1 cells and Nrf2 heterozygous mice. Western blotting, real time PCR, IHC staining and ELISA showed that Nrf2 deficiency could restrain the increase in MTR expression and the decrease in Hcy level induced by vitamin D. The transcriptional activities of Nrf2/MTR were activated by vitamin D/VDR with a decrease in Hcy. CONCLUSION Vitamin D/VDR upregulates MTR in an Nrf2-dependent manner, thereby reducing the risk of HHcy.
Collapse
Affiliation(s)
- Xiaoqi Sun
- Department of Clinical Nutrition, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Ning Liu
- Department of Clinical Nutrition, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Can Sun
- Department of Clinical Nutrition, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Yingxi Xu
- Department of Clinical Nutrition, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Ding Ding
- Department of Clinical Nutrition, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Juan Kong
- Department of Clinical Nutrition, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| |
Collapse
|
11
|
Anitha A, Thanseem I, Iype M, Thomas SV. Mitochondrial dysfunction in cognitive neurodevelopmental disorders: Cause or effect? Mitochondrion 2023; 69:18-32. [PMID: 36621534 DOI: 10.1016/j.mito.2023.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 12/21/2022] [Accepted: 01/04/2023] [Indexed: 01/07/2023]
Abstract
Mitochondria have a crucial role in brain development and neurogenesis, both in embryonic and adult brains. Since the brain is the highest energy consuming organ, it is highly vulnerable to mitochondrial dysfunction. This has been implicated in a range of brain disorders including, neurodevelopmental conditions, psychiatric illnesses, and neurodegenerative diseases. Genetic variations in mitochondrial DNA (mtDNA), and nuclear DNA encoding mitochondrial proteins, have been associated with several cognitive disorders. However, it is not yet clear whether mitochondrial dysfunction is a primary cause of these conditions or a secondary effect. Our review article deals with this topic, and brings out recent advances in mitochondria-oriented therapies. Mitochondrial dysfunction could be involved in the pathogenesis of a subset of disorders involving cognitive impairment. In these patients, mitochondrial dysfunction could be the cause of the condition, rather than the consequence. There are vast areas in this topic that remains to be explored and elucidated.
Collapse
Affiliation(s)
- Ayyappan Anitha
- Dept. of Neurogenetics, Institute for Communicative and Cognitive Neurosciences (ICCONS), Shoranur, Palakkad 679 523, Kerala, India.
| | - Ismail Thanseem
- Dept. of Neurogenetics, Institute for Communicative and Cognitive Neurosciences (ICCONS), Shoranur, Palakkad 679 523, Kerala, India
| | - Mary Iype
- Dept. of Pediatric Neurology, Government Medical College, Thiruvananthapuram 695 011, Kerala, India; Dept. of Neurology, ICCONS, Thiruvananthapuram 695 033, Kerala, India
| | - Sanjeev V Thomas
- Dept. of Neurology, ICCONS, Thiruvananthapuram 695 033, Kerala, India
| |
Collapse
|
12
|
Bagheri L, Jafari-Gharabaghlou D, Valizadeh H, Barzegari A, Zarghami N. Design and development of biodegradable POSS-PCL-Zeolite (β) nano-scaffold for potential applications in bone regeneration. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2023:1-20. [PMID: 36680788 DOI: 10.1080/09205063.2023.2170675] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Side effects caused by bone fractures and restrictions on bone regeneration impose an enormous economic burden on the health system of society. To overcome these limitations, tissue engineering and cell-based therapies have been proposed as alternatives to induce and promote bone healing. Still, bone regeneration disadvantages, such as limited and painful surgery, the risk of infection, nerve injury, bleeding, and function damage, have led investigators to find an alternative therapy. In some studies, bone stimulants have prompted scientists to design scaffolds with appropriate physical structure with the possibility of cell adhesion and proliferation, which plays an influential role in the regeneration and repair of bone tissue. PCL nanofiber is an absorbing candidate for the formulation of biocompatible scaffolds used in tissue engineering. To overcome these negative aspects, improve the properties of PCL nanofibers, and based on the biocompatibility and superior mechanical properties of POSS, Polyhedral Oligomeric Silsesquioxane-Polycaprolactone-Zeolite (POSS-PCL-Zeolite) nanocomposite electrospun nanofiber scaffolds were fabricated in the present study. Nanohybrids and nanofibers structures were characterized by FTIR, HNMR, XRD, SEM, EDX, and DSC techniques. We used cellular and molecular assays, including DCFH ROS detection system, gene expression (RUNX-2, Osteocalcin, Nrf2, BAX, VEGF gens), and apoptotic to demonstrate the biocompatibility and induce bone differentiation of formulated POSS-PCL-Zeolite scaffolds. The results showed the biodegradability of POSS-PCL-Zeolite Nano-scaffold and supported the nesting of mesenchymal stem cells (MSCs) and induced bone differentiation by POSS-PCL-Zeolite Nano-scaffold.
Collapse
Affiliation(s)
- Leyla Bagheri
- Department of Chemistry, Faculty of Sciences, Azarbaijan Shahid Madani University, Tabriz, Iran.,Department of Clinical Biochemistry and Laboratory Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Davoud Jafari-Gharabaghlou
- Department of Clinical Biochemistry and Laboratory Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hasan Valizadeh
- Department of Chemistry, Faculty of Sciences, Azarbaijan Shahid Madani University, Tabriz, Iran
| | - Abolfazl Barzegari
- Research Center for Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nosratollah Zarghami
- Department of Medical Biochemistry, Faculty of Medicine, Istanbul Aydin University, Istanbul, Turkey
| |
Collapse
|
13
|
Wang Y, Dong Q, Hu S, Zou H, Wu T, Shi J, Zhang H, Sheng Y, Sun W, Kong X, Chen L. Decoding microbial genomes to understand their functional roles in human complex diseases. IMETA 2022; 1:e14. [PMID: 38868571 PMCID: PMC10989872 DOI: 10.1002/imt2.14] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 01/20/2022] [Accepted: 02/28/2022] [Indexed: 06/14/2024]
Abstract
Complex diseases such as cardiovascular disease (CVD), obesity, inflammatory bowel disease (IBD), kidney disease, type 2 diabetes (T2D), and cancer have become a major burden to public health and affect more than 20% of the population worldwide. The etiology of complex diseases is not yet clear, but they are traditionally thought to be caused by genetics and environmental factors (e.g., dietary habits), and by their interactions. Besides this, increasing pieces of evidence now highlight that the intestinal microbiota may contribute substantially to the health and disease of the human host via their metabolic molecules. Therefore, decoding the microbial genomes has been an important strategy to shed light on their functional potential. In this review, we summarize the roles of the gut microbiome in complex diseases from its functional perspective. We further introduce artificial tools in decoding microbial genomes to profile their functionalities. Finally, state-of-the-art techniques have been highlighted which may contribute to a mechanistic understanding of the gut microbiome in human complex diseases and promote the development of the gut microbiome-based personalized medicine.
Collapse
Affiliation(s)
- Yifeng Wang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical UniversityNanjing Medical UniversityNanjingJiangsuChina
- Cardiovascular Research Center, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu SchoolNanjing Medical UniversitySuzhouJiangsuChina
| | - Quanbin Dong
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical UniversityNanjing Medical UniversityNanjingJiangsuChina
- Cardiovascular Research Center, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu SchoolNanjing Medical UniversitySuzhouJiangsuChina
| | - Shixian Hu
- Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat‐Sen UniversitySun Yat‐Sen UniversityGuangzhouGuangdongChina
| | - Huayiyang Zou
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical UniversityNanjing Medical UniversityNanjingJiangsuChina
| | - Tingting Wu
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical UniversityNanjing Medical UniversityNanjingJiangsuChina
| | - Jing Shi
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical UniversityNanjing Medical UniversityNanjingJiangsuChina
| | - Haifeng Zhang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical UniversityNanjing Medical UniversityNanjingJiangsuChina
- Cardiovascular Research Center, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu SchoolNanjing Medical UniversitySuzhouJiangsuChina
| | - Yanhui Sheng
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical UniversityNanjing Medical UniversityNanjingJiangsuChina
- Cardiovascular Research Center, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu SchoolNanjing Medical UniversitySuzhouJiangsuChina
| | - Wei Sun
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical UniversityNanjing Medical UniversityNanjingJiangsuChina
- Cardiovascular Research Center, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu SchoolNanjing Medical UniversitySuzhouJiangsuChina
| | - Xiangqing Kong
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical UniversityNanjing Medical UniversityNanjingJiangsuChina
- Cardiovascular Research Center, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu SchoolNanjing Medical UniversitySuzhouJiangsuChina
| | - Lianmin Chen
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical UniversityNanjing Medical UniversityNanjingJiangsuChina
- Cardiovascular Research Center, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu SchoolNanjing Medical UniversitySuzhouJiangsuChina
- Department of Genetics, University Medical Center GroningenUniversity of GroningenGroningenThe Netherlands
| |
Collapse
|
14
|
Liu X, Lin J, Zhang H, Khan NU, Zhang J, Tang X, Cao X, Shen L. Oxidative Stress in Autism Spectrum Disorder-Current Progress of Mechanisms and Biomarkers. Front Psychiatry 2022; 13:813304. [PMID: 35299821 PMCID: PMC8921264 DOI: 10.3389/fpsyt.2022.813304] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 01/24/2022] [Indexed: 12/11/2022] Open
Abstract
Autism spectrum disorder (ASD) is a type of neurodevelopmental disorder that has been diagnosed in an increasing number of children around the world. Existing data suggest that early diagnosis and intervention can improve ASD outcomes. However, the causes of ASD remain complex and unclear, and there are currently no clinical biomarkers for autism spectrum disorder. More mechanisms and biomarkers of autism have been found with the development of advanced technology such as mass spectrometry. Many recent studies have found a link between ASD and elevated oxidative stress, which may play a role in its development. ASD is caused by oxidative stress in several ways, including protein post-translational changes (e.g., carbonylation), abnormal metabolism (e.g., lipid peroxidation), and toxic buildup [e.g., reactive oxygen species (ROS)]. To detect elevated oxidative stress in ASD, various biomarkers have been developed and employed. This article summarizes recent studies about the mechanisms and biomarkers of oxidative stress. Potential biomarkers identified in this study could be used for early diagnosis and evaluation of ASD intervention, as well as to inform and target ASD pharmacological or nutritional treatment interventions.
Collapse
Affiliation(s)
- Xukun Liu
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, China.,Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China.,Shenzhen Key Laboratory of Marine Biotechnology and Ecology, Shenzhen, China
| | - Jing Lin
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, China
| | - Huajie Zhang
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, China
| | - Naseer Ullah Khan
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, China
| | - Jun Zhang
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, China
| | - Xiaoxiao Tang
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, China
| | - Xueshan Cao
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, China
| | - Liming Shen
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, China.,Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China.,Brain Disease and Big Data Research Institute, Shenzhen University, Shenzhen, China
| |
Collapse
|
15
|
An R, Li D, Dong Y, She Q, Zhou T, Nie X, Pan R, Deng Y. Methylcobalamin Protects Melanocytes from H 2O 2-Induced Oxidative Stress by Activating the Nrf2/HO-1 Pathway. Drug Des Devel Ther 2021; 15:4837-4848. [PMID: 34876806 PMCID: PMC8643160 DOI: 10.2147/dddt.s336066] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 11/11/2021] [Indexed: 12/28/2022] Open
Abstract
Purpose Oxidative stress is considered a major determinant in the pathogenesis of vitiligo. Methylcobalamin (MeCbl) is an activated form of vitamin B12 that regulates inflammatory factors, counters oxidative stress, and reduces apoptosis in many disease models. However, the specific mechanism of MeCbl repigmentation against vitiligo is unknown. In this study, we explored the effect of MeCbl on melanocytes following hydrogen peroxide (H2O2)-induced oxidative stress. Methods We established an oxidative stress model using the immortalized human normal melanocyte cell line PIG1. We used a Cell Counting Kit-8 (CCK-8) to detect drug cytotoxicity, and we measured the melanin content of cells using the NaOH method. Intracellular oxidative damage was assessed by flow cytometry and antioxidant enzyme detection kits. In addition, we assessed the presence of apoptosis by flow cytometry and Western blots. We explored the underlying mechanisms of MeCbl during oxidative stress in melanocytes by analyzing the results of experiments based on real-time quantitative polymerase chain reaction (RT-qPCR), Western blotting, and laser scanning confocal immunofluorescence microscopy. Finally, we repeated the experiments after applying an inhibitor to block the Nrf2 pathway. Results We found that MeCbl treatment enhanced cell viability, increased melanin content, reduced intracellular reactive oxygen species (ROS) accumulation, increased the activities of antioxidant enzyme superoxide dismutase (SOD) and catalase (CAT), reduced melanocyte apoptosis, and up-regulated the expression of the Nrf2/HO-1 pathway. Moreover, the protective effects of MeCbl were significantly weakened after inhibiting the Nrf2/HO-1 pathway. Conclusion Our results indicate that MeCbl attenuated the H2O2-induced oxidative stress in melanocytes by activating the Nrf2/HO-1 pathway, this suggests that MeCbl may be an effective treatment against vitiligo.
Collapse
Affiliation(s)
- Ran An
- Department of Dermatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China.,Department of Dermatology, Children's Hospital of Soochow University, Suzhou, Jiangsu, People's Republic of China
| | - Dong Li
- Department of Dermatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Yingying Dong
- Department of Dermatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Qiuyun She
- Department of Dermatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Ting Zhou
- Department of Dermatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Xiaoqi Nie
- Department of Dermatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Ronghua Pan
- Department of Dermatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Yunhua Deng
- Department of Dermatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| |
Collapse
|