1
|
Rodrigues GZP, Finkler M, Dos Santos TG, Kayser JM, Lima DDD, Burghausen JH, de Oliveira DL, Ziulkoski AL, Gehlen G. Chronic Exposure of Zebrafish to Iron and Aluminum: Evaluation of Reversal and Generational Transposition of Behavioral, Histopathological, and Genotoxic Changes. ENVIRONMENTAL TOXICOLOGY 2025; 40:583-597. [PMID: 39575842 DOI: 10.1002/tox.24443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 10/24/2024] [Accepted: 11/11/2024] [Indexed: 03/18/2025]
Abstract
This study aimed to report the effects of chronic exposure of zebrafish exposed to environmentally relevant concentrations of 0.5, 2.4, and 5.0 mg L-1 iron (Fe) and 0.2, 0.4, and 2.0 mg L-1 aluminum (Al). We also evaluated the reversal and generational transposition (F1) of possible histopathological, behavioral, and genotoxic changes in the species. Locomotion changes that may have been caused by the increase in the number of apoptotic cells and in the telencephalic mitochondrial activity were observed especially after the 30 days exposure to Al and persisted after recovery (30 days). We also observed histopathological changes, such as an increase in the number of intestinal goblet cells, even after the recovery period in these animals. Our results also showed that the Fe concentrations used were insufficient to cause genotoxicity, behavioral and intestinal epithelium changes. The adult offspring (F1) of animals exposed to Al showed changes in locomotion and in the amount of goblet cells, demonstrating that even in low concentrations this pollutant can harm subsequent generations in the aquatic biota. Animals demonstrate, in general, greater tolerance to Fe which may be related to the physiological demand of this metal by the body. Even so, all concentrations of both metals that caused some change in the species represent Brazilian environmental occurrences or Brazilian legislation. It highlights the need for updating the guidelines and constant monitoring of aquatic environments, since even in the face of a hypothetical decontamination of the environment, some changes could persist and affect different trophic levels.
Collapse
Affiliation(s)
- Gabriela Zimmermann Prado Rodrigues
- Post Graduation Program in Environmental Quality, Feevale University, Novo Hamburgo, Rio Grande do Sul, Brazil
- Biomedicine Course, CESUCA University Center, Cachoeirinha, Rio Grande do Sul, Brazil
| | - Mariana Finkler
- Post Graduation Program in Environmental Quality, Feevale University, Novo Hamburgo, Rio Grande do Sul, Brazil
| | - Thainá Garbino Dos Santos
- Post Graduation Program in Biological Sciences, Biochemistry, Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Juliana Machado Kayser
- Master's Degree in Toxicology and Toxicological Analysis, Feevale University, Novo Hamburgo, Rio Grande do Sul, Brazil
| | - Diego Del Duca Lima
- Post Graduation Program in Biological Sciences, Biochemistry, Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Jorge Henrique Burghausen
- Post Graduation Program in Environmental Quality, Feevale University, Novo Hamburgo, Rio Grande do Sul, Brazil
| | - Diogo Losch de Oliveira
- Post Graduation Program in Biological Sciences, Biochemistry, Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Ana Luiza Ziulkoski
- Post Graduation Program in Environmental Quality, Feevale University, Novo Hamburgo, Rio Grande do Sul, Brazil
| | - Günther Gehlen
- Post Graduation Program in Environmental Quality, Feevale University, Novo Hamburgo, Rio Grande do Sul, Brazil
| |
Collapse
|
2
|
Zhang Y, Li H, Li B, Li Y, Chai X, Li S, Xue X, Li H, Zhao Y, Tang Y, Yin B, Zhao P, Li E, Feng P. Dachaihu decoction ameliorates abnormal behavior by regulating gut microbiota in rats with propionic acid-induced autism. Front Microbiol 2025; 16:1535451. [PMID: 40018671 PMCID: PMC11867326 DOI: 10.3389/fmicb.2025.1535451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 01/23/2025] [Indexed: 03/01/2025] Open
Abstract
Background Autism spectrum disorder (ASD) is an early-onset neurodevelopmental disorder, usually accompanied by gut microbiota dysregulation. Gut microbiota homeostasis is considered effective for ASD. Reportedly, Dachaihu decoction (DCHD) can efficiently regulate gut microbiota and inflammation. However, the mechanisms underlying the effects of DCHD in the treatment of ASD remain unclear. Objective This study investigated the potential effects and mechanisms of DCHD in treating ASD. Methods In the animal experiment, propionic acid was administered to construct an ASD rat model. The ASD rats were treated with DCHD, and the efficacy was assessed using the behavioral detections, such as open field test, elevated plus maze test, novel object recognition test. Additionally, the levels of IL-6, TNF-α, IL-10, T-SOD, MDA, GSH and CAT were determined using kits, and histological staining was used to evaluate brain morphology. Moreover, tight junction proteins (ZO-1 and occludin) expression levels were evaluated using RT-qPCR, whereas Iba1 expression level was assessed by immunofluorescence staining. The 16S rRNA sequencing and metabolomic analysis of feces revealed the potential targets of DCHD against ASD. In a small human trail, the clinical scales ADOS-2 and Autism Behavior Checklist (ABC) assessed autism severity. Gastrointestinal problems and brain function were evaluated based on food intolerance and event-related potential, respectively. Results DCHD significantly improved autism-like behaviors and increased antioxidant enzyme activity, decreased inflammation and enhanced the intestinal barrier by the animal experiment. Furthermore, the DCHD treatment altered the gut microbiota profile, with increased probiotics Adlercreutzia, Parvibacter, Turicibacter, and Christensenellaceae. Further, DCHD increased the beneficial metabolite indole-3-acetate and decreased the cognitive impairment-related metabolites asymmetric dimethylarginine and homogentisic acid. Meanwhile, the small clinical trial revealed that DCHD significantly alleviated the core symptoms of ASD, with decreased ADOS-2 and ABC scale scores. DCHD also decreased the levels of specific egg white/yolk and milk IgG antibodies and shortened the MMN and P3b latencies. Conclusion This study demonstrated that DCHD may alleviate ASD via inhibiting oxidative stress, reducing inflammation, and modulating the gut microbiota in rats. Combined with human trial, DCHD may be a promising drug for treating ASD. This study provides a scientific rationale for treating mental disorders related to gut microbiota dysbiosis.
Collapse
Affiliation(s)
- Yangyang Zhang
- Department of Children Rehabilitation, Henan Key Laboratory of Rehabilitation Medicine, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hang Li
- Henan Key Laboratory for Helicobacter Pylori and Digestive Tract Microecology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Bolin Li
- College of Traditional Chinese Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Yizhuang Li
- Department of Neurology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xuejun Chai
- School of Basic Medical Science, The Shaanxi Key Laboratory of Brain Disorders, Xi’an Medical University, Xi’an, China
| | - Sheng Li
- School of Life Sciences, Westlake University, Hangzhou, China
| | - Xia Xue
- Henan Key Laboratory for Helicobacter Pylori and Digestive Tract Microecology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Honglei Li
- Department of Children Rehabilitation, Henan Key Laboratory of Rehabilitation Medicine, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yonghong Zhao
- Department of Children Rehabilitation, Henan Key Laboratory of Rehabilitation Medicine, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Youcai Tang
- Department of Children Rehabilitation, Henan Key Laboratory of Rehabilitation Medicine, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Provincial Outstanding Overseas Scientists Chronic Liver Injury Workshop, Zhengzhou Key Laboratory of Metabolism-Associated Fatty Liver Disease, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Baoqi Yin
- Department of Children Rehabilitation, Henan Key Laboratory of Rehabilitation Medicine, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Pengju Zhao
- Department of Children Rehabilitation, Henan Key Laboratory of Rehabilitation Medicine, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Enyao Li
- Department of Children Rehabilitation, Henan Key Laboratory of Rehabilitation Medicine, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Pengya Feng
- Department of Children Rehabilitation, Henan Key Laboratory of Rehabilitation Medicine, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory for Helicobacter Pylori and Digestive Tract Microecology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
3
|
Flores Ventura E, Bernabeu M, Callejón-Leblic B, Cabrera-Rubio R, Yeruva L, Estañ-Capell J, Martínez-Costa C, García-Barrera T, Collado MC. Human milk metals and metalloids shape infant microbiota. Food Funct 2024; 15:12134-12145. [PMID: 39584920 DOI: 10.1039/d4fo01929f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2024]
Abstract
Background: The profile of metal(loid)s in human milk is essential for infant growth and development, yet its impact on the development of the infant microbiota remains unclear. Elements, such as manganese, zinc, iron or copper, play crucial roles in influencing infant health. Aim: To investigate the metal(loid) content within human milk and its influence on the infant's gut microbiota within the first 2 months after birth. Methods: Human milk samples and infant stool samples from 77 mother-infant dyads in the MAMI cohort were collected at two time points: the early transitional stage and the mature stage. Metallomic profiling of human milk was conducted using inductively coupled plasma-mass spectrometry (ICP-MS). The infant gut microbiota was profiled through 16S rRNA amplicon sequencing and maternal-infant clinical data were available. Spearman's rank correlation coefficientsprovided insights into metal(loid)-microbiota relationships. Results: Independent cross-sectional analyses of mother-infant pairs at two time points, significant variations in metal concentrations and differences in microbial abundances and diversities were observed. Notably, Bifidobacterium genus abundance was higher during the mature lactation stage. During early lactation, we found a significant positive correlation between infant gut Corynebacterium and human milk nickel concentrations, and negative correlations between Veillonella spp. and antimony, and Enterobacter spp. and copper. Additionally, Simpson's diversity was negatively correlated with iron. In the mature lactation stage, we identified eleven significant correlations between metals and microbiota. Notably, Klebsiella genus showed multiple negative correlations with iron, antimony, and vanadium. Conclusion: Our study highlights the significance of metal(loid)-microbiota interactions in early infant development, indicating that infant gut Klebsiella genus may be particularly vulnerable to fluctuations in metal(loid) levels present in human milk, when compared to other genera. Future research should explore these interactions at a strain level and the implications on infant health and development. This trial was registered as NCT03552939.
Collapse
Affiliation(s)
- Eduard Flores Ventura
- Institute of Agrochemistry and Food Technology-National Research Council (IATA-CSIC), Valencia, Spain.
| | - Manuel Bernabeu
- Institute of Agrochemistry and Food Technology-National Research Council (IATA-CSIC), Valencia, Spain.
| | - Belén Callejón-Leblic
- Research Centre of Natural Resources, Health and the Environment (RENSMA), Department of Chemistry, Faculty of Experimental Sciences, University of Huelva, Campus El Carmen, Fuerzas Armadas Ave., 21007, Huelva, Spain
| | - Raúl Cabrera-Rubio
- Institute of Agrochemistry and Food Technology-National Research Council (IATA-CSIC), Valencia, Spain.
| | - Laxmi Yeruva
- Microbiome and Metabolism Research Unit, USDA-ARS, SEA, Arkansas Children's Nutrition Center, Little Rock, AR, USA
| | - Javier Estañ-Capell
- Department of Pediatrics, University of Valencia, INCLIVA Biomedical Research Institute, Avenida Blasco Ibáñez 15-17, 46010 Valencia, Spain
| | - Cecilia Martínez-Costa
- Department of Pediatrics, University of Valencia, INCLIVA Biomedical Research Institute, Avenida Blasco Ibáñez 15-17, 46010 Valencia, Spain
| | - Tamara García-Barrera
- Research Centre of Natural Resources, Health and the Environment (RENSMA), Department of Chemistry, Faculty of Experimental Sciences, University of Huelva, Campus El Carmen, Fuerzas Armadas Ave., 21007, Huelva, Spain
| | - María Carmen Collado
- Institute of Agrochemistry and Food Technology-National Research Council (IATA-CSIC), Valencia, Spain.
| |
Collapse
|
4
|
Cheng H, Liu J, Zhang D, Wu J, Wu J, Zhou Y, Tan Y, Feng W, Peng C. Natural products: Harnessing the power of gut microbiota for neurological health. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156019. [PMID: 39305747 DOI: 10.1016/j.phymed.2024.156019] [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: 02/25/2024] [Revised: 05/19/2024] [Accepted: 09/01/2024] [Indexed: 12/01/2024]
Abstract
BACKGROUND Neurological diseases are the primary cause of disability and death and impose substantial financial burdens. However, existing treatments only relieve symptoms and may cause many adverse effects. Natural products are a promising source of neurological therapeutic agents due to their excellent neuroprotective effect and safety. The gut microbiota has an essential impact on maintaining brain homeostasis via the gut-brain axis. Multiple investigations show that natural products offer neuroprotective effects by regulating gut microbiota-driven signaling networks. OBJECTIVES This review aims to provide a systematic review of how natural products promote neurological health by harnessing the power of gut microbiota. METHODS The pre-January 1, 2024 literature was gathered from several databases, including Scopus, PubMed, Google Scholar, and Web of Science, utilizing appropriate keywords. The gathered publications underwent a review process and were classified based on their study content, specifically focusing on the impact of natural products on gut microbiota and neurological health. RESULTS Here, we review how natural products promote neurological health by regulating the gut microbiota-brain axis. Specifically, we focus on the following areas. (1) Altering microorganism community structure, including increasing α-diversity and altering β-diversity. (2) Regulating the population of certain bacteria, including enriching beneficial microorganisms Akkermansia and Bifidobacterium, and inhibiting potentially hazardous microorganisms Bilophila, Klebsiella, and Helicobacter. (3) Regulating microbial neuroactive metabolites levels, including short-chain fatty acids, tryptophan and its derivatives, trimethylamine N-oxide, dopa/dopamine, γ-aminobutyric acid, and lipopolysaccharide. Furthermore, we review how natural products promote neurological health by regulating intestinal barrier homeostasis. CONCLUSION Natural products promote neurological health by harnessing the power of gut microbiota. This review will contribute to understanding how natural products promote neurological health by orchestrating the gut microbiota-brain axis.
Collapse
Affiliation(s)
- Hao Cheng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Juan Liu
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Dandan Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jing Wu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jinlu Wu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yaochuan Zhou
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Yuzhu Tan
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Wuwen Feng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Cheng Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China; School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| |
Collapse
|
5
|
Tavares-Silva E, de Aquino Lemos V, de França E, Silvestre J, dos Santos SA, Ravacci GR, Thomatieli-Santos RV. Protective Effects of Probiotics on Runners' Mood: Immunometabolic Mechanisms Post-Exercise. Nutrients 2024; 16:3761. [PMID: 39519594 PMCID: PMC11547471 DOI: 10.3390/nu16213761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Revised: 10/24/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND The gut-brain axis may mediate mood changes due to strenuous exercise. Therefore, probiotic supplementation may mitigate mood worsening. PURPOSE The present study aims to evaluate the effect of probiotic supplementation on mood and immunometabolic parameters after a marathon. MATERIALS AND METHODS Fourteen marathon runners were selected and divided into placebo and probiotic groups that were supplemented for 30 days. Before and after the marathon, mood (POMS) was assessed, and blood was collected for analysis of immunometabolic parameters. Statistical analysis was performed, and p < 0.05 was considered to determine statistically differences. RESULTS Tension decreased after the marathon in both groups. Vigor decreased only in the placebo group. Fatigue increased after the marathon in both groups. TMD increased after the marathon in placebo. The IL2/IL-4 ratio decreased in the probiotic group after the marathon compared to before and increased compared to the placebo group. The IL-10 increased after the marathon in placebo. TNF-α increased after the marathon in probiotics. The TNF-α/IL-10 ratio decreased after the marathon in both groups. LPS decreased in the probiotic group after the marathon compared to before and in the placebo group. CONCLUSIONS Thirty days of probiotic supplementation attenuated the impact of marathons on mood worsening. The decrease in LPS in the probiotic group mediated the change in the pro/anti-inflammatory balance, indicating an immunometabolic mechanism by which the gut-brain axis impacts mood after strenuous exercise.
Collapse
Affiliation(s)
- Edgar Tavares-Silva
- Post-Graduate Program in Psychobiology, Federal University of São Paulo, São Paulo 04040-003, SP, Brazil (V.d.A.L.)
| | - Valdir de Aquino Lemos
- Post-Graduate Program in Psychobiology, Federal University of São Paulo, São Paulo 04040-003, SP, Brazil (V.d.A.L.)
| | - Elias de França
- Department of Bioscience, Federal University of São Paulo, Rua Silva Jardim, 136, Vila Mathias, Santos 11015-021, SP, Brazil; (E.d.F.); (J.S.); (S.A.d.S.); (G.R.R.)
| | - Jean Silvestre
- Department of Bioscience, Federal University of São Paulo, Rua Silva Jardim, 136, Vila Mathias, Santos 11015-021, SP, Brazil; (E.d.F.); (J.S.); (S.A.d.S.); (G.R.R.)
| | - Samile Amorim dos Santos
- Department of Bioscience, Federal University of São Paulo, Rua Silva Jardim, 136, Vila Mathias, Santos 11015-021, SP, Brazil; (E.d.F.); (J.S.); (S.A.d.S.); (G.R.R.)
| | - Graziela Rosa Ravacci
- Department of Bioscience, Federal University of São Paulo, Rua Silva Jardim, 136, Vila Mathias, Santos 11015-021, SP, Brazil; (E.d.F.); (J.S.); (S.A.d.S.); (G.R.R.)
| | - Ronaldo Vagner Thomatieli-Santos
- Post-Graduate Program in Psychobiology, Federal University of São Paulo, São Paulo 04040-003, SP, Brazil (V.d.A.L.)
- Department of Bioscience, Federal University of São Paulo, Rua Silva Jardim, 136, Vila Mathias, Santos 11015-021, SP, Brazil; (E.d.F.); (J.S.); (S.A.d.S.); (G.R.R.)
| |
Collapse
|
6
|
Li L, Fu M, Chen F, Ji H, Zhou G, Chen L, Geng H, Guo J, Pei L, Sun J. The mediating effect of circulating inflammatory proteins on the relationship between gut microbiota and FD: a bidirectional Mendelian randomization study. Sci Rep 2024; 14:23785. [PMID: 39390038 PMCID: PMC11466956 DOI: 10.1038/s41598-024-74717-6] [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: 05/23/2024] [Accepted: 09/27/2024] [Indexed: 10/12/2024] Open
Abstract
Functional dyspepsia (FD) is known to be influenced by gut microbiota (GM) and circulating inflammatory proteins (CIPs), however, the causal relationship between GM, CIPs and FD haven't been investigated. This study employed two-sample Mendelian Randomization (TSMR) to investigate their associations using data from Genome-Wide Association Studies (GWAS). In this study, Inverse-variance weighted (IVW) method was employed as the primary analysis, with supplementary approaches including weighted median, weighted mode, simple mode, and MR-Egger. Heterogeneity and pleiotropy were assessed using the Cochrane Q test, MR-Egger intercept test, and MR-PRESSO global test. Totally, 196 GM traits and 91 CIPs were analyzed, and the results uncovered the causal impact of 12 GM taxa and 5 proteins on functional dyspepsia (FD). 9 GM genera were linked to a reduced risk of FD, while 3 GM genera were associated with an increased risk of FD.Additionally, reverse analysis revealed no FD-GM causation. Furthermore, IL-12, IL-10, CXCL10, CXCL9 and VEGFA were significantly correlated with FD, with CXCL9 and VEGFA acting as mediators in the association between GM traits and FD. Taken together, our findings established a link between specific GM and CIPs in the pathogenesis of FD, offering novel insights for its diagnosis and treatment.
Collapse
Affiliation(s)
- Li Li
- Department of Acupuncture and Rehabilitation, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
| | - Minhan Fu
- Department of Acupuncture and Rehabilitation, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
| | - Feiyi Chen
- Department of Acupuncture and Rehabilitation, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
| | - Huijie Ji
- Department of Ophthalmology, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
| | - Guowei Zhou
- Department of General Surgery, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
| | - Lu Chen
- Department of Acupuncture and Rehabilitation, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
| | - Hao Geng
- Department of Acupuncture and Rehabilitation, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
| | - Jing Guo
- College of Health and Rehabilitation, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Lixia Pei
- Department of Acupuncture and Rehabilitation, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China.
| | - Jianhua Sun
- Department of Acupuncture and Rehabilitation, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China.
| |
Collapse
|
7
|
Cepeda Y, Elizondo-Vega R, Garrido C, Tobar C, Araneda M, Oliveros P, Ordenes P, Carril C, Vidal PM, Luz-Crawford P, García-Robles MA, Oyarce K. Regulatory T cells administration reduces anxiety-like behavior in mice submitted to chronic restraint stress. Front Cell Neurosci 2024; 18:1406832. [PMID: 39206016 PMCID: PMC11349540 DOI: 10.3389/fncel.2024.1406832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 07/25/2024] [Indexed: 09/04/2024] Open
Abstract
Background Major depression disorder (MDD) and anxiety are common mental disorders that significantly affect the quality of life of those who suffer from them, altering the person's normal functioning. From the biological perspective, the most classical hypothesis explaining their occurrence relies on neurotransmission and hippocampal excitability alterations. However, around 30% of MDD patients do not respond to medication targeting these processes. Over the last decade, the involvement of inflammatory responses in depression and anxiety pathogenesis has been strongly acknowledged, opening the possibility of tackling these disorders from an immunological point of view. In this context, regulatory T cells (Treg cells), which naturally maintain immune homeostasis by suppressing inflammation could be promising candidates for their therapeutic use in mental disorders. Methods To test this hypothesis, C57BL/6 adult male mice were submitted to classical stress protocols to induce depressive and anxiety-like behavior; chronic restriction stress (CRS), and chronic unpredictable stress (CUS). Some of the stressed mice received a single adoptive transfer of Treg cells during stress protocols. Mouse behavior was analyzed through the open field (OFT) and forced swim test (FST). Blood and spleen samples were collected for T cell analysis using cell cytometry, while brains were collected to study changes in microglia by immunohistochemistry. Results Mice submitted to CRS and CUS develop anxiety and depressive-like behavior, and only CRS mice exhibit lower frequencies of circulating Treg cells. Adoptive transfer of Treg cells decreased anxiety-like behavior in the OFT only in CRS model, but not depressive behavior in FST in neither of the two models. In CRS mice, Treg cells administration lowered the number of microglia in the hippocampus, which increased due this stress paradigm, and restored its arborization. However, in CUS mice, Treg cells administration increased microglia number with no significant effect on their arborization. Conclusion Our results for effector CD4+ T cells in the spleen and microglia number and morphology in the hippocampus add new evidence in favor of the participation of inflammatory responses in the development of depressive and anxiety-like behavior and suggest that the modulation of key immune cells such as Treg cells, could have beneficial effects on these disorders.
Collapse
Affiliation(s)
- Yamila Cepeda
- Laboratorio de Neuroinmunología, Facultad de Medicina y Ciencia, Universidad San Sebastián, Sede Concepción, Concepción, Chile
- Laboratorio de Biología Celular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Roberto Elizondo-Vega
- Laboratorio de Biología Celular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Camila Garrido
- Laboratorio de Neuroinmunología, Facultad de Medicina y Ciencia, Universidad San Sebastián, Sede Concepción, Concepción, Chile
- Laboratorio de Biología Celular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Catalina Tobar
- Laboratorio de Neuroinmunología, Facultad de Medicina y Ciencia, Universidad San Sebastián, Sede Concepción, Concepción, Chile
| | - Matías Araneda
- Laboratorio de Neuroinmunología, Facultad de Medicina y Ciencia, Universidad San Sebastián, Sede Concepción, Concepción, Chile
| | - Patricia Oliveros
- Laboratorio de Neuroinmunología, Facultad de Medicina y Ciencia, Universidad San Sebastián, Sede Concepción, Concepción, Chile
| | - Patricio Ordenes
- Laboratorio de Neuroinmunología, Facultad de Medicina y Ciencia, Universidad San Sebastián, Sede Concepción, Concepción, Chile
| | - Claudio Carril
- Laboratorio de Neuroinmunología, Facultad de Medicina y Ciencia, Universidad San Sebastián, Sede Concepción, Concepción, Chile
| | - Pía M. Vidal
- Neuroimmunology and Regeneration of the Central Nervous System Unit, Biomedical Science Research Laboratory, Department of Basic Sciences, Faculty of Medicine, Universidad Católica de la Santísima Concepción, Concepción, Chile
| | - Patricia Luz-Crawford
- Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - María. A. García-Robles
- Laboratorio de Biología Celular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Karina Oyarce
- Laboratorio de Neuroinmunología, Facultad de Medicina y Ciencia, Universidad San Sebastián, Sede Concepción, Concepción, Chile
| |
Collapse
|
8
|
Frileux S, Boltri M, Doré J, Leboyer M, Roux P. Cognition and gut microbiota in schizophrenia spectrum and mood disorders: A systematic review. Neurosci Biobehav Rev 2024; 162:105722. [PMID: 38754717 DOI: 10.1016/j.neubiorev.2024.105722] [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: 02/01/2024] [Revised: 04/11/2024] [Accepted: 05/10/2024] [Indexed: 05/18/2024]
Abstract
FRILEUX, M., BOLTRI M. and al. Cognition and Gut microbiota in schizophrenia spectrum and mood disorders: a Systematic Review. NEUROSCI BIOBEHAV REV (1) 2024 Schizophrenia spectrum disorders and major mood disorders are associated with cognitive impairments. Recent studies suggest a link between gut microbiota composition and cognitive functioning. Here, we review the relationship between gut microbiota and cognition in these disorders. To do this, we conducted a systematic review, searching Cochrane Central Register of Controlled Trials, EBSCOhost, Embase, Pubmed, Scopus, and Web of Science. Studies were included if they investigated the relationship between gut microbiota composition and cognitive function through neuropsychological assessments in patients with bipolar, depressive, schizophrenia spectrum, and other psychotic disorders. Ten studies were identified. Findings underscore a link between gut dysbiosis and cognitive impairment. This relationship identified specific taxa (Haemophilus, Bacteroides, and Alistipes) as potential contributors to bolstered cognitive performance. Conversely, Candida albicans, Toxoplasma gondii, Streptococcus and Deinococcus were associated with diminished performance on cognitive assessments. Prebiotics and probiotics interventions were associated with cognitive enhancements, particularly executive functions. These results emphasize the role of gut microbiota in cognition, prompting further exploration of the underlying mechanisms paving the way toward precision psychiatry.
Collapse
Affiliation(s)
- S Frileux
- Service Hospitalo-Universitaire de Psychiatrie d'Adultes et d'Addictologie, Centre Hospitalier de Versailles, 177, rue de Versailles, Le Chesnay-Rocquencourt 78157, France; Université Paris-Saclay, Université Versailles Saint-Quentin-En-Yvelines, DisAP-DevPsy-CESP, INSERM UMR1018, Villejuif 94807, France.
| | - M Boltri
- Department of Psychology, Catholic University of Sacred Heart, Milan, Italy; I.R.C.C.S. Istituto Auxologico Italiano, Experimental Laboratory for Metabolic Neurosciences Research, Piancavallo, Italy
| | - J Doré
- Université Paris-Saclay, INRA, MetaGenoPolis, AgroParisTech, MICALIS, Jouy-en-Josas 78350, France
| | - M Leboyer
- Inserm U955 IMRB, Translational Neuropsychiatry Laboratory, AP-HP, DMU IMPACT, Fédération Hospitalo-Universitaire de médecine de précision en psychiatrie (FHU ADAPT), Paris Est Créteil University and Fondation FondaMental, Créteil 94010, France; Fondation Fondamental, Créteil 94010, France
| | - P Roux
- Service Hospitalo-Universitaire de Psychiatrie d'Adultes et d'Addictologie, Centre Hospitalier de Versailles, 177, rue de Versailles, Le Chesnay-Rocquencourt 78157, France; Université Paris-Saclay, Université Versailles Saint-Quentin-En-Yvelines, DisAP-DevPsy-CESP, INSERM UMR1018, Villejuif 94807, France
| |
Collapse
|
9
|
Liu P, Liu Z, Wang J, Wang J, Gao M, Zhang Y, Yang C, Zhang A, Li G, Li X, Liu S, Liu L, Sun N, Zhang K. Immunoregulatory role of the gut microbiota in inflammatory depression. Nat Commun 2024; 15:3003. [PMID: 38589368 PMCID: PMC11001948 DOI: 10.1038/s41467-024-47273-w] [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: 07/09/2023] [Accepted: 03/26/2024] [Indexed: 04/10/2024] Open
Abstract
Inflammatory depression is a treatment-resistant subtype of depression. A causal role of the gut microbiota as a source of low-grade inflammation remains unclear. Here, as part of an observational trial, we first analyze the gut microbiota composition in the stool, inflammatory factors and short-chain fatty acids (SCFAs) in plasma, and inflammatory and permeability markers in the intestinal mucosa of patients with inflammatory depression (ChiCTR1900025175). Gut microbiota of patients with inflammatory depression exhibits higher Bacteroides and lower Clostridium, with an increase in SCFA-producing species with abnormal butanoate metabolism. We then perform fecal microbiota transplantation (FMT) and probiotic supplementation in animal experiments to determine the causal role of the gut microbiota in inflammatory depression. After FMT, the gut microbiota of the inflammatory depression group shows increased peripheral and central inflammatory factors and intestinal mucosal permeability in recipient mice with depressive and anxiety-like behaviors. Clostridium butyricum administration normalizes the gut microbiota, decreases inflammatory factors, and displays antidepressant-like effects in a mouse model of inflammatory depression. These findings suggest that inflammatory processes derived from the gut microbiota can be involved in neuroinflammation of inflammatory depression.
Collapse
Affiliation(s)
- Penghong Liu
- Department of Psychiatry, First Hospital of Shanxi Medical University, Taiyuan, 030001, PR China
- Shanxi Medical University, Taiyuan, 030001, PR China
- Shanxi Key Laboratory of Artificial Intelligence Assisted Diagnosis and Treatment for Mental Disorder, First Hospital of Shanxi Medical University, Taiyuan, 030001, PR China
| | - Zhifen Liu
- Department of Psychiatry, First Hospital of Shanxi Medical University, Taiyuan, 030001, PR China
- Shanxi Key Laboratory of Artificial Intelligence Assisted Diagnosis and Treatment for Mental Disorder, First Hospital of Shanxi Medical University, Taiyuan, 030001, PR China
- Key Laboratory of Cellular Physiology (Shanxi Medical University), Ministry of Education, Taiyuan, 030001, PR China
| | - Jizhi Wang
- Department of Psychiatry, First Hospital of Shanxi Medical University, Taiyuan, 030001, PR China
- Shanxi Medical University, Taiyuan, 030001, PR China
| | - Junyan Wang
- Department of Psychiatry, First Hospital of Shanxi Medical University, Taiyuan, 030001, PR China
- Shanxi Medical University, Taiyuan, 030001, PR China
| | - Mingxue Gao
- Department of Psychiatry, First Hospital of Shanxi Medical University, Taiyuan, 030001, PR China
- Shanxi Medical University, Taiyuan, 030001, PR China
| | - Yanyan Zhang
- Department of Psychiatry, First Hospital of Shanxi Medical University, Taiyuan, 030001, PR China
- Shanxi Medical University, Taiyuan, 030001, PR China
| | - Chunxia Yang
- Department of Psychiatry, First Hospital of Shanxi Medical University, Taiyuan, 030001, PR China
- Shanxi Key Laboratory of Artificial Intelligence Assisted Diagnosis and Treatment for Mental Disorder, First Hospital of Shanxi Medical University, Taiyuan, 030001, PR China
| | - Aixia Zhang
- Department of Psychiatry, First Hospital of Shanxi Medical University, Taiyuan, 030001, PR China
- Shanxi Key Laboratory of Artificial Intelligence Assisted Diagnosis and Treatment for Mental Disorder, First Hospital of Shanxi Medical University, Taiyuan, 030001, PR China
| | - Gaizhi Li
- Department of Psychiatry, First Hospital of Shanxi Medical University, Taiyuan, 030001, PR China
- Shanxi Medical University, Taiyuan, 030001, PR China
- Shanxi Key Laboratory of Artificial Intelligence Assisted Diagnosis and Treatment for Mental Disorder, First Hospital of Shanxi Medical University, Taiyuan, 030001, PR China
| | - Xinrong Li
- Department of Psychiatry, First Hospital of Shanxi Medical University, Taiyuan, 030001, PR China
- Shanxi Key Laboratory of Artificial Intelligence Assisted Diagnosis and Treatment for Mental Disorder, First Hospital of Shanxi Medical University, Taiyuan, 030001, PR China
| | - Sha Liu
- Department of Psychiatry, First Hospital of Shanxi Medical University, Taiyuan, 030001, PR China
- Shanxi Medical University, Taiyuan, 030001, PR China
- Shanxi Key Laboratory of Artificial Intelligence Assisted Diagnosis and Treatment for Mental Disorder, First Hospital of Shanxi Medical University, Taiyuan, 030001, PR China
| | - Lixin Liu
- Experimental Center of Science and Research, First Hospital of Shanxi Medical University, Taiyuan, 030001, PR China
| | - Ning Sun
- Department of Psychiatry, First Hospital of Shanxi Medical University, Taiyuan, 030001, PR China.
- Shanxi Medical University, Taiyuan, 030001, PR China.
- Shanxi Key Laboratory of Artificial Intelligence Assisted Diagnosis and Treatment for Mental Disorder, First Hospital of Shanxi Medical University, Taiyuan, 030001, PR China.
| | - Kerang Zhang
- Department of Psychiatry, First Hospital of Shanxi Medical University, Taiyuan, 030001, PR China.
- Shanxi Medical University, Taiyuan, 030001, PR China.
- Shanxi Key Laboratory of Artificial Intelligence Assisted Diagnosis and Treatment for Mental Disorder, First Hospital of Shanxi Medical University, Taiyuan, 030001, PR China.
| |
Collapse
|
10
|
Tinkov AA, Skalny AV, Domingo JL, Samarghandian S, Kirichuk AA, Aschner M. A review of the epidemiological and laboratory evidence of the role of aluminum exposure in pathogenesis of cardiovascular diseases. ENVIRONMENTAL RESEARCH 2024; 242:117740. [PMID: 38007081 DOI: 10.1016/j.envres.2023.117740] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 11/13/2023] [Accepted: 11/18/2023] [Indexed: 11/27/2023]
Abstract
The objective of the present study was to review the epidemiological and laboratory evidence on the role of aluminum (Al) exposure in the pathogenesis of cardiovascular diseases. Epidemiological data demonstrated an increased incidence of cardiovascular diseases (CVD), including hypertension and atherosclerosis in occupationally exposed subjects and hemodialysis patients. In addition, Al body burden was found to be elevated in patients with coronary heart disease, hypertension, and dyslipidemia. Laboratory studies demonstrated that Al exposure induced significant ultrastructural damage in the heart, resulting in electrocardiogram alterations in association with cardiomyocyte necrosis and apoptosis, inflammation, oxidative stress, inflammation, and mitochondrial dysfunction. In agreement with the epidemiological findings, laboratory data demonstrated dyslipidemia upon Al exposure, resulting from impaired hepatic lipid catabolism, as well as promotion of low-density lipoprotein oxidation. Al was also shown to inhibit paraoxonase 1 activity and to induce endothelial dysfunction and adhesion molecule expression, further promoting atherogenesis. The role of Al in hypertension was shown to be mediated by up-regulation of NADPH-oxidase, inhibition of nitric oxide bioavailability, and stimulation of renin-angiotensin-aldosterone system. It has been also demonstrated that Al exposure targets cerebral vasculature, which may be considered a link between Al exposure and cerebrovascular diseases. Findings from other tissues lend support that ferroptosis, pyroptosis, endoplasmic reticulum stress, and modulation of gut microbiome and metabolome are involved in the development of CVD upon Al exposure. A better understanding of the role of the cardiovascular system as a target for Al toxicity will be useful for risk assessment and the development of treatment and prevention strategies.
Collapse
Affiliation(s)
- Alexey A Tinkov
- Center of Bioelementology and Human Ecology, IM Sechenov First Moscow State Medical University (Sechenov University), Moscow, 119435, Russia; Laboratory of Ecobiomonitoring and Quality Control, Yaroslavl State University, Yaroslavl, 150003, Russia; Department of Human Ecology and Bioelementology, and Department of Medical Elementology, Peoples' Friendship University of Russia (RUDN University), Moscow, 117198, Russia.
| | - Anatoly V Skalny
- Center of Bioelementology and Human Ecology, IM Sechenov First Moscow State Medical University (Sechenov University), Moscow, 119435, Russia; Department of Human Ecology and Bioelementology, and Department of Medical Elementology, Peoples' Friendship University of Russia (RUDN University), Moscow, 117198, Russia
| | - Jose L Domingo
- Laboratory of Toxicology and Environmental Health, School of Medicine, Universitat Rovira I Virgili, 4320, Reus, Catalonia, Spain
| | - Saeed Samarghandian
- Healthy Ageing Research Centre, Neyshabur University of Medical Sciences, Neyshabur, 9319774446, Iran
| | - Anatoly A Kirichuk
- Department of Human Ecology and Bioelementology, and Department of Medical Elementology, Peoples' Friendship University of Russia (RUDN University), Moscow, 117198, Russia
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| |
Collapse
|
11
|
Wu M, Chen C, Lei H, Cao Z, Zhang C, Du R, Zhang C, Song Y, Qin M, Zhou J, Lu Y, Wang X, Zhang L. Dietary Isoquercetin Ameliorates Bone Loss via Restoration of the Gut Microbiota and Lipopolysaccharide-Triggered Inflammatory Status in Ovariectomy Mice. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:15981-15990. [PMID: 37852299 DOI: 10.1021/acs.jafc.3c00205] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/20/2023]
Abstract
Osteoporosis is one of the skeletal degenerative diseases accompanied by bone loss and microstructure disruption. Given that the gut-bone signaling axis highly contributes to bone health, here, dietary isoquercetin (IQ) was shown to effectively improve postmenopausal osteoporosis (PMO) in an ovariectomy (OVX) mouse model through the modulation of the gut-bone cross-talk. An in vivo study showed that OVX induced striking disruption of the microbial community, subsequently causing gut leakage and gut barrier dysfunction. As a result, lipopolysaccharide (LPS)-triggered inflammatory cytokines released from the intestine to bone marrow were determined to be associated with bone loss in OVX mice. Long-term dietary IQ effectively improved microbial community and gut barrier function in the OVX mice and thus markedly improved bone loss and host inflammatory status by repressing the NF-κB signaling pathway. An in vitro study further revealed that IQ treatments dose-dependently inhibited LPS-induced inflammation and partly promoted the proliferation and differentiation of osteoblasts. These results provide new evidence that dietary IQ has the potential for osteoporosis treatment.
Collapse
Affiliation(s)
- Mengjing Wu
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Centre for Magnetic Resonance in Wuhan, Innovation Academy for Precision Measurement Science and Technology, CAS, Wuhan 430071, China
- College of Chemistry and Materials Science, South-Central Minzu University, Wuhan 430074, China
| | - Chuan Chen
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Centre for Magnetic Resonance in Wuhan, Innovation Academy for Precision Measurement Science and Technology, CAS, Wuhan 430071, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Hehua Lei
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Centre for Magnetic Resonance in Wuhan, Innovation Academy for Precision Measurement Science and Technology, CAS, Wuhan 430071, China
| | - Zheng Cao
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Centre for Magnetic Resonance in Wuhan, Innovation Academy for Precision Measurement Science and Technology, CAS, Wuhan 430071, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Cui Zhang
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Centre for Magnetic Resonance in Wuhan, Innovation Academy for Precision Measurement Science and Technology, CAS, Wuhan 430071, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ruichen Du
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Centre for Magnetic Resonance in Wuhan, Innovation Academy for Precision Measurement Science and Technology, CAS, Wuhan 430071, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ce Zhang
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Centre for Magnetic Resonance in Wuhan, Innovation Academy for Precision Measurement Science and Technology, CAS, Wuhan 430071, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yuchen Song
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Centre for Magnetic Resonance in Wuhan, Innovation Academy for Precision Measurement Science and Technology, CAS, Wuhan 430071, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Mengyu Qin
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Centre for Magnetic Resonance in Wuhan, Innovation Academy for Precision Measurement Science and Technology, CAS, Wuhan 430071, China
- College of Chemistry and Materials Science, South-Central Minzu University, Wuhan 430074, China
| | - Jinlin Zhou
- Golden Health (Guangdong) Biotechnology Co., Ltd, Foshan 528225, China
- Engineering Research Academy of High Value Utilization of Green Plants, Meizhou 514021, China
| | - Yujing Lu
- Golden Health (Guangdong) Biotechnology Co., Ltd, Foshan 528225, China
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Xian Wang
- College of Chemistry and Materials Science, South-Central Minzu University, Wuhan 430074, China
| | - Limin Zhang
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Centre for Magnetic Resonance in Wuhan, Innovation Academy for Precision Measurement Science and Technology, CAS, Wuhan 430071, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
12
|
Maeda N, Yumoto T, Xiong G, Hasegawa Y. Characterization and Stability of a Novel Toxin in Scallop Mantle Tissue. Foods 2023; 12:3224. [PMID: 37685157 PMCID: PMC10487249 DOI: 10.3390/foods12173224] [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: 07/20/2023] [Revised: 08/21/2023] [Accepted: 08/25/2023] [Indexed: 09/10/2023] Open
Abstract
Previous studies have shown that mice fed a diet containing 1% mantle tissue exhibited decreased food consumption and led to death. Toxic substances present in the mantle tissue have been isolated and identified. In the present study, we explored the characteristics and stability of mantle tissue toxicity. The treatment of mantle tissue with 1 mM hydrochloric acid, 1 mM sodium hydroxide, 1 mM dithiothreitol, and 1 mM hydrogen peroxide followed by heating did not significantly reduce the toxicity of mantle tissue in mice. These results suggest that mantle toxins are stable in tissues, particularly when exposed to acidic conditions and digestive enzymes. We examined whether mantle tissue exhibited acute toxicity. Mice fed a diet containing 20% mantle tissue did not show a distinct increase in toxicity compared with mice fed a diet containing 1% mantle tissue, demonstrating that feeding mantle tissue does not lead to acute toxicity. Finally, mantle tissue toxicity in the small intestine was examined. Chronic feeding of mantle tissue to mice changed the color of the small intestine. Real-time polymerase chain reaction analysis revealed that mantle tissue feeding caused changes in inflammation and endoplasmic reticulum stress markers in the small intestine. These results suggest that mantle tissue feeding causes toxicity after initial damage to the small intestinal tissue.
Collapse
Affiliation(s)
| | | | | | - Yasushi Hasegawa
- College of Environmental Technology, Muroran Institute of Technology, 27-1 Mizumoto, Muroran 050-8585, Japan; (N.M.); (T.Y.); (G.X.)
| |
Collapse
|
13
|
Chen W, Du L, Cai C, Huang L, Zheng Q, Chen J, Wang L, Zhang X, Fang X, Wang L, Zhong Q, Zhong W, Wang J, Liao Z. Take chicks as an example: Rummeliibacillus stabekisii CY2 enhances immunity and regulates intestinal microbiota by degrading LPS to promote organism growth and development. J Funct Foods 2023; 105:105583. [DOI: 10.1016/j.jff.2023.105583] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025] Open
|
14
|
Sasso J, Ammar RM, Tenchov R, Lemmel S, Kelber O, Grieswelle M, Zhou QA. Gut Microbiome-Brain Alliance: A Landscape View into Mental and Gastrointestinal Health and Disorders. ACS Chem Neurosci 2023; 14:1717-1763. [PMID: 37156006 PMCID: PMC10197139 DOI: 10.1021/acschemneuro.3c00127] [Citation(s) in RCA: 65] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 04/24/2023] [Indexed: 05/10/2023] Open
Abstract
Gut microbiota includes a vast collection of microorganisms residing within the gastrointestinal tract. It is broadly recognized that the gut and brain are in constant bidirectional communication, of which gut microbiota and its metabolic production are a major component, and form the so-called gut microbiome-brain axis. Disturbances of microbiota homeostasis caused by imbalance in their functional composition and metabolic activities, known as dysbiosis, cause dysregulation of these pathways and trigger changes in the blood-brain barrier permeability, thereby causing pathological malfunctions, including neurological and functional gastrointestinal disorders. In turn, the brain can affect the structure and function of gut microbiota through the autonomic nervous system by regulating gut motility, intestinal transit and secretion, and gut permeability. Here, we examine data from the CAS Content Collection, the largest collection of published scientific information, and analyze the publication landscape of recent research. We review the advances in knowledge related to the human gut microbiome, its complexity and functionality, its communication with the central nervous system, and the effect of the gut microbiome-brain axis on mental and gut health. We discuss correlations between gut microbiota composition and various diseases, specifically gastrointestinal and mental disorders. We also explore gut microbiota metabolites with regard to their impact on the brain and gut function and associated diseases. Finally, we assess clinical applications of gut-microbiota-related substances and metabolites with their development pipelines. We hope this review can serve as a useful resource in understanding the current knowledge on this emerging field in an effort to further solving of the remaining challenges and fulfilling its potential.
Collapse
Affiliation(s)
- Janet
M. Sasso
- CAS, a division of the American Chemical Society, 2540 Olentangy River Rd, Columbus, Ohio 43202, United States
| | - Ramy M. Ammar
- Bayer
Consumer Health, R&D Digestive
Health, Darmstadt 64295, Germany
| | - Rumiana Tenchov
- CAS, a division of the American Chemical Society, 2540 Olentangy River Rd, Columbus, Ohio 43202, United States
| | - Steven Lemmel
- CAS, a division of the American Chemical Society, 2540 Olentangy River Rd, Columbus, Ohio 43202, United States
| | - Olaf Kelber
- Bayer
Consumer Health, R&D Digestive
Health, Darmstadt 64295, Germany
| | - Malte Grieswelle
- Bayer
Consumer Health, R&D Digestive
Health, Darmstadt 64295, Germany
| | - Qiongqiong Angela Zhou
- CAS, a division of the American Chemical Society, 2540 Olentangy River Rd, Columbus, Ohio 43202, United States
| |
Collapse
|
15
|
Chen Z, Yang H, Fu H, Wu L, Liu M, Jiang H, Liu Q, Wang Y, Xiong S, Zhou M, Sun X, Chen C, Huang L. Gut bacterial species in late trimester of pregnant sows influence the occurrence of stillborn piglet through pro-inflammation response. Front Immunol 2023; 13:1101130. [PMID: 36741405 PMCID: PMC9890068 DOI: 10.3389/fimmu.2022.1101130] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 12/28/2022] [Indexed: 01/19/2023] Open
Abstract
Maternal gut microbiota is an important regulator for the metabolism and immunity of the fetus during pregnancy. Recent studies have indicated that maternal intestinal microbiota is closely linked to the development of fetus and infant health. Some bacterial metabolites are considered to be directly involved in immunoregulation of fetus during pregnancy. However, the detailed mechanisms are largely unknown. In this study, we exploited the potential correlation between the gut microbiota of pregnant sows and the occurrence of stillborn piglets by combining the 16S rRNA gene and metagenomic sequencing data, and fecal metabolome in different cohorts. The results showed that several bacterial species from Bacteroides, potential pathogens, and LPS-producing bacteria exhibited significantly higher abundances in the gut of sows giving birth to stillborn piglets. Especially, Bacteroides fragilis stood out as the key driver in both tested cohorts and showed the most significant association with the occurrence of stillborn piglets in the DN1 cohort. However, several species producing short-chain fatty acids (SCFAs), such as Prevotella copri, Clostridium butyricum and Faecalibacterium prausnitzii were enriched in the gut of normal sows. Functional capacity analysis of gut microbiome revealed that the pathways associated with infectious diseases and immune diseases were enriched in sows giving birth to stillborn piglets. However, energy metabolism had higher abundance in normal sows. Fecal metabolome profiling analysis found that Lysophosphatidylethanolamine and phosphatidylethanolamine which are the main components of cell membrane of Gram-negative bacteria showed significantly higher concentration in stillbirth sows, while SCFAs had higher concentration in normal sows. These metabolites were significantly associated with the stillborn-associated bacterial species including Bacteroides fragilis. Lipopolysaccharide (LPS), IL-1β, IL-6, FABP2, and zonulin had higher concentration in the serum of stillbirth sows, indicating increased intestinal permeability and pro-inflammatory response. The results from this study suggested that certain sow gut bacterial species in late trimester of pregnancy, e.g., an excess abundance of Bacteroides fragilis, produced high concentration of LPS which induced sow pro-inflammatory response and might cause the death of the relatively weak piglets in a farrow. This study provided novel evidences about the effect of maternal gut microbiota on the fetus development and health.
Collapse
Affiliation(s)
| | - Hui Yang
- *Correspondence: Lusheng Huang, ; Congying Chen, ; Hui Yang,
| | | | | | | | | | | | | | | | | | | | - Congying Chen
- *Correspondence: Lusheng Huang, ; Congying Chen, ; Hui Yang,
| | - Lusheng Huang
- *Correspondence: Lusheng Huang, ; Congying Chen, ; Hui Yang,
| |
Collapse
|
16
|
Zhang X, Shen M, Wang C, Gao M, Wang L, Jin Z, Xia X. Impact of aluminum exposure on oxidative stress, intestinal changes and immune responses in red swamp crayfish (Procambarus clarkii). THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 855:158902. [PMID: 36152855 DOI: 10.1016/j.scitotenv.2022.158902] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 07/12/2022] [Accepted: 09/17/2022] [Indexed: 06/16/2023]
Abstract
Aluminum (Al) is an abundant metal that has been classified as a threatening pollutant due to indiscriminate use and anthropogenic activities. This study aimed to evaluate the impacts of Al on crayfish (Procambarus clarkii), including biochemical change, histological alteration, gut microbial community diversification, and immune changes. The bioaccumulation of Al was detected in the hemolymph and intestine of crayfish after Al exposure at different time points. Results showed that Al exposure significantly induced oxidative stress and caused pathohistological changes on intestinal barrier structures in crayfish. It was found that the intestinal microbiota was affected by retained Al and the intestinal community diversity was changed after Al treated in the crayfish. Furthermore, Al exposure affected the immunity in crayfish, by altering the expression of a set of immune-related genes, as well as reducing the phenoloxidase and lysozyme activities. Moreover, Al exposure promoted hemocytes apoptosis and impaired hemophagocytic capacity against Vibro parahamolyticus, resulting in higher mortality of crayfish upon bacterial infection. Taken these results together, we conclude that excessive Al exposure caused adverse effects on multiple biological processes of crayfish and Al pollution is a potential threat to crayfish culture.
Collapse
Affiliation(s)
- Xiaowen Zhang
- College of Life Sciences, Henan Normal University, Xinxiang, Henan 453007, China; Henan International Joint Laboratory of Agricultural Microbial Ecology and Technology, Henan Normal University, Xinxiang 453007, China; The Observation and Research Field Station of Taihang Mountain Forest Ecosystems of Henan Province, Xinxiang 453007, Henan, China
| | - Manli Shen
- College of Life Sciences, Henan Normal University, Xinxiang, Henan 453007, China
| | - Cui Wang
- College of Life Sciences, Henan Normal University, Xinxiang, Henan 453007, China
| | - Miaomiao Gao
- College of Life Sciences, Henan Normal University, Xinxiang, Henan 453007, China
| | - Liuen Wang
- College of Life Sciences, Henan Normal University, Xinxiang, Henan 453007, China
| | - Zeyu Jin
- College of Life Sciences, Henan Normal University, Xinxiang, Henan 453007, China
| | - Xiaohua Xia
- College of Life Sciences, Henan Normal University, Xinxiang, Henan 453007, China; The Observation and Research Field Station of Taihang Mountain Forest Ecosystems of Henan Province, Xinxiang 453007, Henan, China.
| |
Collapse
|
17
|
Alterations of the Composition and Neurometabolic Profile of Human Gut Microbiota in Major Depressive Disorder. Biomedicines 2022; 10:biomedicines10092162. [PMID: 36140263 PMCID: PMC9496097 DOI: 10.3390/biomedicines10092162] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 08/27/2022] [Accepted: 08/29/2022] [Indexed: 12/21/2022] Open
Abstract
Major depressive disorder (MDD) is among the most prevalent mental disorders worldwide. Factors causing the pathogenesis of MDD include gut microbiota (GM), which interacts with the host through the gut–brain axis. In previous studies of GM in MDD patients, 16S rRNA sequencing was used, which provided information about composition but not about function. In our study, we analyzed whole metagenome sequencing data to assess changes in both the composition and functional profile of GM. We looked at the GM of 36 MDD patients, compared with that of 38 healthy volunteers. Comparative taxonomic analysis showed decreased abundances of Faecalibacterium prausnitzii, Roseburia hominis, and Roseburia intestinalis, and elevated abundances of Escherichia coli and Ruthenibacterium lactatiformans in the GM of MDD patients. We observed decreased levels of bacterial genes encoding key enzymes involved in the production of arginine, asparagine, glutamate, glutamine, melatonin, acetic, butyric and conjugated linoleic acids, and spermidine in MDD patients. These genes produced signature pairs with Faecalibacterium prausntizii and correlated with decreased levels of this species in the GM of MDD patients. These results show the potential impact of the identified biomarker bacteria and their metabolites on the pathogenesis of MDD, and should be confirmed in future metabolomic studies.
Collapse
|
18
|
Zhou L, Zeng Y, Zhang H, Ma Y. The Role of Gastrointestinal Microbiota in Functional Dyspepsia: A Review. Front Physiol 2022; 13:910568. [PMID: 35755434 PMCID: PMC9214042 DOI: 10.3389/fphys.2022.910568] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 05/23/2022] [Indexed: 12/18/2022] Open
Abstract
Functional dyspepsia is a clinically common functional gastrointestinal disorder with a high prevalence, high impact and high consumption of medical resources. The microbiota in the gastrointestinal tract is a large number of families and is one of the most complex microbial reservoirs in the human body. An increasing number of studies have confirmed the close association between dysbiosis of the gastrointestinal microbiota and the occurrence and progression of functional dyspepsia. Therefore, we reviewed the role of dysbiosis of the gastrointestinal microbiota, H. pylori infection and gastrointestinal microbiota metabolites in functional dyspepsia, focusing on the possible mechanisms by which dysbiosis of the gastrointestinal microbiota contributes to the pathogenesis of functional dyspepsia. Several studies have confirmed that dysbiosis of the gastrointestinal microbiota may cause the occurrence and progression of functional dyspepsia by disrupting the biological barrier of the intestinal mucosa, by disturbing the immune function of the intestinal mucosa, or by causing dysregulation of the microbial-gut-brain axis. Probiotics and antibiotics have also been chosen to treat functional dyspepsia in clinical studies and have shown some improvement in the clinical symptoms. However, more studies are needed to explore and confirm the relationship between dysbiosis of the gastrointestinal microbiota and the occurrence and progression of functional dyspepsia, and more clinical studies are needed to confirm the therapeutic efficacy of microbiota modulation for functional dyspepsia.
Collapse
Affiliation(s)
- Li Zhou
- Department of Rehabilitation Medicine, Wuhan Hospital of Integrated Traditional Chinese and Western Medicine, Wuhan, China
| | - Yi Zeng
- Department of Hospital Infection Management Office, Wuhan Hospital of Integrated Traditional Chinese and Western Medicine, Wuhan, China
| | - Hongxing Zhang
- Department of Acupuncture, Wuhan Hospital of Integrated Traditional Chinese and Western Medicine, Wuhan, China
| | - Yan Ma
- Department of Rehabilitation Medicine, Wuhan Hospital of Integrated Traditional Chinese and Western Medicine, Wuhan, China
| |
Collapse
|
19
|
Hao W, Zhu X, Liu Z, Song Y, Wu S, Lu X, Yang J, Jin C. Resveratrol alleviates aluminum-induced intestinal barrier dysfunction in mice. ENVIRONMENTAL TOXICOLOGY 2022; 37:1373-1381. [PMID: 35156769 DOI: 10.1002/tox.23490] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 01/27/2022] [Accepted: 02/05/2022] [Indexed: 06/14/2023]
Abstract
BACKGROUND Aluminum is mainly exposed to the general population through food and water, and is absorbed into the systemic circulation through intestine, which in turn damages the intestinal barrier. METHODS The mice model of subchronic exposure to aluminum chloride (AlCl3 ) was established via oral. Tail suspension test was used to detect depressive behavior. H&E staining was performed to assess pathological intestinal injury. Intestinal permeability was estimated by exogenous Evans blue content. The level of inflammatory cytokines and tight junction protein were assessed via ELISA and western blotting. Simultaneously, resveratrol (Rsv, an agonist of Sirt1) was evaluated the protective role against intestinal barrier injuries caused by aluminum exposure. RESULTS Our results showed that AlCl3 induced depressive-like behavior, intestinal pathological damage and intestinal barrier permeability, resulting in intestinal barrier dysfunction. Besides, aluminum induced the expression of inflammatory cytokines, which further triggered IRF8-MMP9-mediated downregulation of tight junction proteins including CLD1, OCLD and ZO-1. After Rsv treatment, SIRT1 expression was increased, depressive symptom was improved, pathological injury was reduced, inflammatory reaction was alleviated, and intestinal barrier function restored. CONCLUSION Our findings revealed that aluminum exposure induced intestinal barrier dysfunction by IRF8-MMP9 signaling pathway. Rsv alleviated these injuries via activating SIRT1.
Collapse
Affiliation(s)
- Wudi Hao
- Department of Toxicology, School of Public Health, China Medical University, Shenyang, China
| | - Xiaoying Zhu
- Department of Toxicology, School of Public Health, China Medical University, Shenyang, China
| | - Ziyue Liu
- Department of Toxicology, School of Public Health, China Medical University, Shenyang, China
| | - Yushuai Song
- Department of Toxicology, School of Public Health, China Medical University, Shenyang, China
| | - Shengwen Wu
- Department of Toxicology, School of Public Health, China Medical University, Shenyang, China
| | - Xiaobo Lu
- Department of Toxicology, School of Public Health, China Medical University, Shenyang, China
| | - Jinghua Yang
- Department of Toxicology, School of Public Health, China Medical University, Shenyang, China
| | - Cuihong Jin
- Department of Toxicology, School of Public Health, China Medical University, Shenyang, China
| |
Collapse
|
20
|
Korf JM, Ganesh BP, McCullough LD. Gut dysbiosis and age-related neurological diseases in females. Neurobiol Dis 2022; 168:105695. [PMID: 35307514 PMCID: PMC9631958 DOI: 10.1016/j.nbd.2022.105695] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Revised: 02/19/2022] [Accepted: 03/13/2022] [Indexed: 12/12/2022] Open
Abstract
Historically, females have been underrepresented in biological research. With increased interest in the gut microbiome and the gut-brain axis, it is important for researchers to pursue studies that consider sex as a biological variable. The composition of the gut microbiome is influenced by environmental factors, disease, diet, and varies with age and by sex. Detrimental changes in the gut microbiome, referred to as dysbiosis, is believed to influence the development and progression of age-related neurodegenerative disorders such as Alzheimer’s disease, Parkinson’s disease, Huntington’s disease, and stroke. Many are investigating the changes in microbial populations in order or to better understand the role of the gut immunity and the microbiome in neurodegenerative diseases, many of which the exact etiology remains elusive, and no cures exist. Others are working to find diagnostic markers for earlier detection, or to therapeutically modulate microbial populations using probiotics. However, while all these diseases present in reproductively senescent females, most studies only use male animals for their experimental design. Reproductively senescent females have been shown to have differences in disease progression, inflammatory responses, and microbiota composition, therefore, for research to be translational to affected populations it is necessary for appropriate models to be used. This review discusses factors that influence the gut microbiome and the gut brain axis in females, and highlights studies that have investigated the role of dysbiosis in age-related neurodegenerative disorders that have included females in their study design.
Collapse
Affiliation(s)
- Janelle M Korf
- Department of Neurology, McGovern Medical School, University of Texas Health Science Center at Houston, 6431 Fannin Street, Houston, TX 77370, USA; University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, 6767 Bertner Ave, Houston, TX 77030, USA.
| | - Bhanu P Ganesh
- Department of Neurology, McGovern Medical School, University of Texas Health Science Center at Houston, 6431 Fannin Street, Houston, TX 77370, USA.
| | - Louise D McCullough
- Department of Neurology, McGovern Medical School, University of Texas Health Science Center at Houston, 6431 Fannin Street, Houston, TX 77370, USA.
| |
Collapse
|
21
|
Hao W, Hao C, Wu C, Xu Y, Jin C. Aluminum induced intestinal dysfunction via mechanical, immune, chemical and biological barriers. CHEMOSPHERE 2022; 288:132556. [PMID: 34648793 DOI: 10.1016/j.chemosphere.2021.132556] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 09/18/2021] [Accepted: 10/10/2021] [Indexed: 06/13/2023]
Abstract
Aluminum is the most abundant metal element in the Earth's crust, which exists naturally in the form of aluminum compounds. Aluminum is mainly absorbed through the gastrointestinal tract, which varies with different aluminum compounds. During this process, aluminum could induce the disruption of intestinal mucosa barrier. However, its underlying mechanism has not been elucidated yet. Previous studies have reported that aluminum can firstly promote the apoptosis of intestinal epithelial cells, destroy the structure of tight-junction proteins, and increase the intestinal permeability, injuring the mechanical barrier of gut. Also, it can induce the activation of immune cells to secrete inflammatory factors, and trigger immune responses, interfering with immune barrier. Moreover, aluminum treatment can regulate intestinal composition and bio-enzyme activity, impairing the function of chemical barrier. In addition, aluminum accumulation can induce an imbalance of the intestinal flora, inhibit the growth of beneficial bacteria, and promote the proliferation of harmful bacteria, which ultimately disrupting biological barrier. Collectively, aluminum may do extensive damage to intestinal barrier function covering mechanical barrier, immune barrier, chemical barrier and biological barrier.
Collapse
Affiliation(s)
- Wudi Hao
- Department of Toxicology, School of Public Health, China Medical University, Shenyang, 110122, PR China
| | - Chenyu Hao
- Department of Toxicology, School of Public Health, China Medical University, Shenyang, 110122, PR China
| | - Chengrong Wu
- Department of Toxicology, School of Public Health, China Medical University, Shenyang, 110122, PR China
| | - Yuqing Xu
- Department of Toxicology, School of Public Health, China Medical University, Shenyang, 110122, PR China
| | - Cuihong Jin
- Department of Toxicology, School of Public Health, China Medical University, Shenyang, 110122, PR China.
| |
Collapse
|
22
|
Liu P, Gao M, Liu Z, Zhang Y, Tu H, Lei L, Wu P, Zhang A, Yang C, Li G, Sun N, Zhang K. Gut Microbiome Composition Linked to Inflammatory Factors and Cognitive Functions in First-Episode, Drug-Naive Major Depressive Disorder Patients. Front Neurosci 2022; 15:800764. [PMID: 35153660 PMCID: PMC8831735 DOI: 10.3389/fnins.2021.800764] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Accepted: 12/30/2021] [Indexed: 01/04/2023] Open
Abstract
OBJECTIVE The microbiota-gut-brain axis, especially the inflammatory pathway, may play a critical role in the pathogenesis of cognitive impairment in major depressive disorder (MDD). However, studies on the microbiota-inflammatory-cognitive function axis in MDD are lacking. The aim of the present study was to analyze the gut microbiota composition and explore the correlation between gut microbiota and inflammatory factors, cognitive function in MDD patients. METHOD Study participants included 66 first-episode, drug naïve MDD patients as well as 43 healthy subjects (HCs). The composition of fecal microbiota was evaluated using16S rRNA sequencing and bioinformatics analysis. The cytokines such as hs-CRP, IL-1β, IL-6, IL-10, and TNF-α in peripheral blood were detected via enzyme linked immunosorbent assay (ELISA); assessment of cognitive functions was performed using the Color Trail Test (CTT), The Repeatable Battery for the Assessment of Neuropsychological Status (RBANS) and the Stroop Color-Word Test (SCWT). RESULTS We found that compared with HCs, MDD patients had cognitive impairments and showed different α-diversity and β-diversity of gut microbiota composition. LDA effect size (LEfSe) analysis found MDD have higher Deinococcaceae and lower Bacteroidaceae, Turicibacteraceae, Clostridiaceae and Barnesiellaceae at family level. Deinococcus and Odoribacter was higher in the MDD group, however, Bacteroides, Alistipes, Turicibacter, Clostridium, Roseburia, and Enterobacter were lower at genus level. Furthermore, In MDD patients, the Bacteroidaceae and Bacteroides were both positively correlated with hsCRP, CCT1, CCT2. Alistipes was positively correlated with IL-6, Word time, Color time, Word-Color time, Color-Word time and negatively correlated with Delayed Memory, Total score and Standardized score. Turicibacteraceae and Turicibacter were both negatively correlated with IL-1β and IL-6. CONCLUSION The present findings confirm that the gut microbiota in MDD patients have altered gut microbes that are closely associated with inflammatory factors and cognitive function in MDD patients.
Collapse
Affiliation(s)
- Penghong Liu
- First Hospital of Shanxi Medical University, Taiyuan, China
| | - Mingxue Gao
- Department of Psychiatry, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Zhifen Liu
- Department of Psychiatry, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Yanyan Zhang
- Department of Psychiatry, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Hongwei Tu
- Department of Psychiatry, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Lei Lei
- Department of Psychiatry, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Peiyi Wu
- Department of Psychiatry, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Aixia Zhang
- Department of Psychiatry, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Chunxia Yang
- Department of Psychiatry, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Gaizhi Li
- Department of Psychiatry, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Ning Sun
- Department of Psychiatry, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Kerang Zhang
- Department of Psychiatry, First Hospital of Shanxi Medical University, Taiyuan, China
| |
Collapse
|
23
|
Peng X, Luo Z, He S, Zhang L, Li Y. Blood-Brain Barrier Disruption by Lipopolysaccharide and Sepsis-Associated Encephalopathy. Front Cell Infect Microbiol 2021; 11:768108. [PMID: 34804998 PMCID: PMC8599158 DOI: 10.3389/fcimb.2021.768108] [Citation(s) in RCA: 128] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 10/14/2021] [Indexed: 12/29/2022] Open
Abstract
As a complex multicellular structure of the vascular system at the central nervous system (CNS), the blood-brain barrier (BBB) separates the CNS from the system circulation and regulates the influx and efflux of substances to maintain the steady-state environment of the CNS. Lipopolysaccharide (LPS), the cell wall component of Gram-negative bacteria, can damage the barrier function of BBB and further promote the occurrence and development of sepsis-associated encephalopathy (SAE). Here, we conduct a literature review of the direct and indirect damage mechanisms of LPS to BBB and the relationship between these processes and SAE. We believe that after LPS destroys BBB, a large number of inflammatory factors and neurotoxins will enter and damage the brain tissue, which will activate brain immune cells to mediate inflammatory response and in turn further destroys BBB. This vicious circle will ultimately lead to the progression of SAE. Finally, we present a succinct overview of the treatment of SAE by restoring the BBB barrier function and summarize novel opportunities in controlling the progression of SAE by targeting the BBB.
Collapse
Affiliation(s)
- Xiaoyao Peng
- Department of Clinical Medicine, School of Clinical Medicine, Southwest Medical University, Luzhou, China
| | - Zhixuan Luo
- Department of Clinical Medicine, School of Clinical Medicine, Southwest Medical University, Luzhou, China
| | - Shuang He
- Department of Clinical Medicine, School of Clinical Medicine, Southwest Medical University, Luzhou, China
| | - Luhua Zhang
- Department of Pathogenic Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Ying Li
- Department of Immunology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| |
Collapse
|
24
|
Hao W, Hao C, Wu C, Xu Y, Wu S, Lu X, Yang J, Jin C. Aluminum impairs cognitive function by activating DDX3X-NLRP3-mediated pyroptosis signaling pathway. Food Chem Toxicol 2021; 157:112591. [PMID: 34614429 DOI: 10.1016/j.fct.2021.112591] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 09/29/2021] [Accepted: 10/01/2021] [Indexed: 11/16/2022]
Abstract
INTRODUCTION Aluminum is a kind of chemical contaminants in food which can induce neurotoxicity. Aluminum exposure is closely related to neurodegenerative diseases (ND), in which neuroinflammation might involve. However, the molecular mechanism of aluminum-induced neuroinflammation through pyroptosis is not fully clarified yet. MATERIAL AND METHODS The mice model of subacute exposure to aluminum chloride (AlCl3) was established. BV2 microglia cells was treated with AlCl3 in vitro. Resveratrol (Rsv) was adopted as intervention agent. RESULTS Our results showed that aluminum induced cognitive impairment, destroying blood brain barrier (BBB), and causing nerve injury in mice. Meanwhile, aluminum could stimulate nucleotide oligomerization domain-like receptor family pyrin domain containing protein 3 (NLRP3) inflammasome assembly and activate caspase-1 (CASP1), inducing gasdermin D (GSDMD)-mediated pyroptosis signaling, releasing cytokines IL-1β and IL-18, further promoting the activation of glial cells to magnify neuroinflammatory response. Moreover, DEAD-box helicase 3 X-linked (DDX3X) and stress granule RasGAP SH3-domain-binding protein 1 (G3BP1) both participated in neuroinflammation induced by aluminum. When co-treated with Rsv, these injuries were alleviated to some extent. CONCLUSION Aluminum exposure could induce nerve cell pyroptosis and neuroinflammation by DDX3X-NLRP3 inflammasome signaling pathway, which could be rescued via Rsv activating sirtuin 1 (SIRT1).
Collapse
Affiliation(s)
- Wudi Hao
- Department of Toxicology, School of Public Health, China Medical University, Shenyang, 110122, PR China
| | - Chenyu Hao
- Department of Toxicology, School of Public Health, China Medical University, Shenyang, 110122, PR China
| | - Chengrong Wu
- Department of Toxicology, School of Public Health, China Medical University, Shenyang, 110122, PR China
| | - Yuqing Xu
- Department of Toxicology, School of Public Health, China Medical University, Shenyang, 110122, PR China
| | - Shengwen Wu
- Department of Toxicology, School of Public Health, China Medical University, Shenyang, 110122, PR China
| | - Xiaobo Lu
- Department of Toxicology, School of Public Health, China Medical University, Shenyang, 110122, PR China
| | - Jinghua Yang
- Department of Toxicology, School of Public Health, China Medical University, Shenyang, 110122, PR China
| | - Cuihong Jin
- Department of Toxicology, School of Public Health, China Medical University, Shenyang, 110122, PR China.
| |
Collapse
|
25
|
Wang J, He M, Guo W, Zhang Y, Sui X, Lin J, Liu X, Li H, Li J, Yang Q, Kan M, Zhang Z, Ming S, Qu X, Li N. Microbiome-Metabolomics Reveals Endogenous Alterations of Energy Metabolism by the Dushen Tang to Attenuate D-Galactose-Induced Memory Impairment in Rats. BIOMED RESEARCH INTERNATIONAL 2021; 2021:6649085. [PMID: 34136571 PMCID: PMC8175156 DOI: 10.1155/2021/6649085] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 04/23/2021] [Accepted: 05/12/2021] [Indexed: 02/07/2023]
Abstract
Aging affects the brain function in elderly individuals, and Dushen Tang (DST) is widely used for the treatment of senile diseases. In this study, the protective effect of DST against memory impairment was evaluated through the Morris water maze (MWM) test and transmission electron microscopy (TEM). A joint analysis was also performed using LC-MS metabolomics and the microbiome. The MWM test showed that DST could significantly improve the spatial memory and learning abilities of rats with memory impairment, and the TEM analysis showed that DST could reduce neuronal damage in the hippocampus of rats with memory impairment. Ten potential biomarkers involving pyruvate metabolism, the synthesis and degradation of ketone bodies, and other metabolic pathways were identified by the metabolomic analysis, and it was found that 3-hydroxybutyric acid and lactic acid were involved in the activation of cAMP signaling pathways. The 16S rDNA sequencing results showed that DST could regulate the structure of the gut microbiota in rats with memory impairment, and these effects were manifested as changes in energy metabolism. These findings suggest that DST exerts a good therapeutic effect on rats with memory impairment and that this effect might be mainly achieved by improving energy metabolism. These findings might lead to the potential development of DST as a drug for the treatment of rats with memory impairment.
Collapse
Affiliation(s)
- Jifeng Wang
- Jilin Provincial Key Laboratory of Biomacromolecules of Chinese Medicine, Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, Jilin 130021, China
| | - Min He
- Jilin Provincial Key Laboratory of Biomacromolecules of Chinese Medicine, Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, Jilin 130021, China
- Leiden University—European Center for Chinese Medicine, Leiden University, 2333CC Leiden, Netherlands
| | - Wenjun Guo
- Jilin Provincial Key Laboratory of Biomacromolecules of Chinese Medicine, Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, Jilin 130021, China
| | - Yanhong Zhang
- Jilin Provincial Key Laboratory of Biomacromolecules of Chinese Medicine, Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, Jilin 130021, China
| | - Xin Sui
- Jilin Provincial Key Laboratory of Biomacromolecules of Chinese Medicine, Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, Jilin 130021, China
| | - Jianan Lin
- Jilin Provincial Key Laboratory of Biomacromolecules of Chinese Medicine, Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, Jilin 130021, China
| | - Xiaoran Liu
- Jilin Provincial Key Laboratory of Biomacromolecules of Chinese Medicine, Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, Jilin 130021, China
| | - Hui Li
- Qian Wei Hospital of Jilin Province, Changchun, Jilin 130117, China
| | - Jing Li
- Jilin Provincial Key Laboratory of Biomacromolecules of Chinese Medicine, Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, Jilin 130021, China
| | - Qing Yang
- Jilin Provincial Key Laboratory of Biomacromolecules of Chinese Medicine, Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, Jilin 130021, China
| | - Mo Kan
- Jilin Provincial Key Laboratory of Biomacromolecules of Chinese Medicine, Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, Jilin 130021, China
| | - Zhuang Zhang
- Jilin Provincial Key Laboratory of Biomacromolecules of Chinese Medicine, Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, Jilin 130021, China
| | - Sitong Ming
- Jilin Provincial Key Laboratory of Biomacromolecules of Chinese Medicine, Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, Jilin 130021, China
| | - Xiaobo Qu
- Jilin Provincial Key Laboratory of Biomacromolecules of Chinese Medicine, Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, Jilin 130021, China
| | - Na Li
- Jilin Provincial Key Laboratory of Biomacromolecules of Chinese Medicine, Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, Jilin 130021, China
| |
Collapse
|
26
|
Maigoro AY, Lee S. Gut Microbiome-Based Analysis of Lipid A Biosynthesis in Individuals with Autism Spectrum Disorder: An In Silico Evaluation. Nutrients 2021; 13:nu13020688. [PMID: 33669978 PMCID: PMC7924848 DOI: 10.3390/nu13020688] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 02/14/2021] [Accepted: 02/18/2021] [Indexed: 12/20/2022] Open
Abstract
The link between autism spectrum disorder (ASD) and the gut microbiome has received much attention, with special focus on gut–brain-axis immunological imbalances. Gastrointestinal problems are one of the major symptoms of ASD and are thought to be related to immune dysregulation. Therefore, in silico analysis was performed on mined data from 36 individuals with ASD and 21 control subjects, with an emphasis on lipid A endotoxin-producing bacteria and their lipopolysaccharide (LPS) metabolic pathways. Analysis of enzyme distribution among the 15 most abundant genera in both groups revealed that almost all these genera utilized five early-stage enzymes responsible for catalyzing the nine conserved lipid A synthesis steps. However, Haemophilus and Escherichia, which were significantly more abundant in individuals with ASD than in the control subjects, possess a complete set of essential lipid A synthesis enzymes. Furthermore, the 10 genera with the greatest increase in individuals with ASD showed high potential for producing late-stage lipid A products. Collectively, these results suggested that the synthesis rate of immunogenic LPS end products is likely to increase in individuals with ASD, which may be related to their gastrointestinal symptoms and elevated inflammatory conditions.
Collapse
|
27
|
Fock EM, Parnova RG. Protective Effect of Mitochondria-Targeted Antioxidants against Inflammatory Response to Lipopolysaccharide Challenge: A Review. Pharmaceutics 2021; 13:pharmaceutics13020144. [PMID: 33499252 PMCID: PMC7910823 DOI: 10.3390/pharmaceutics13020144] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 01/15/2021] [Accepted: 01/17/2021] [Indexed: 12/16/2022] Open
Abstract
Lipopolysaccharide (LPS), the major component of the outer membrane of Gram-negative bacteria, is the most abundant proinflammatory agent. Considerable evidence indicates that LPS challenge inescapably causes oxidative stress and mitochondrial dysfunction, leading to cell and tissue damage. Increased mitochondrial reactive oxygen species (mtROS) generation triggered by LPS is known to play a key role in the progression of the inflammatory response. mtROS at excessive levels impair electron transport chain functioning, reduce the mitochondrial membrane potential, and initiate lipid peroxidation and oxidative damage of mitochondrial proteins and mtDNA. Over the past 20 years, a large number of mitochondria-targeted antioxidants (mito-AOX) of different structures that can accumulate inside mitochondria and scavenge free radicals have been synthesized. Their protective role based on the prevention of oxidative stress and the restoration of mitochondrial function has been demonstrated in a variety of common diseases and pathological states. This paper reviews the current data on the beneficial application of different mito-AOX in animal endotoxemia models, in either in vivo or in vitro experiments. The results presented in our review demonstrate the promising potential of approaches based on mito-AOX in the development of new treatment strategies against Gram-negative infections and LPS per se.
Collapse
|
28
|
Zhao Y, Jaber V, Lukiw WJ. Gastrointestinal Tract Microbiome-Derived Pro-inflammatory Neurotoxins in Alzheimer's Disease. JOURNAL OF AGING SCIENCE 2021; 9:002. [PMID: 34671696 PMCID: PMC8525708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The microbiome contained within the human gastrointestinal (GI)-tract constitutes a highly complex, dynamic and interactive internal prokaryotic ecosystem that possesses a staggering diversity, speciation and complexity. This repository of microbes comprises the largest interactive source and highest density of microbes anywhere in nature, collectively constituting the largest 'diffuse organ system' in the human body. Through the extracellular fluid (ECF), cerebrospinal fluid (CSF), lymphatic and glymphatic circulation, endocrine, systemic and neurovascular circulation and/or central and peripheral nervous systems (CNS, PNS) microbiome-derived signaling strongly impacts the health, well-being and vitality of the human host. Recent data from the Human Microbiome Initiative (HMI) and the Unified Human Gastrointestinal Genome (UHGG) consortium have classified over ~200 thousand diverse, non-redundant prokaryotic genomes in the human GI-tract microbiome involving about ~5 thousand different GI-tract microbes that all together encode almost ~200 million different protein sequences. While the largest proportion of different microbiome-derived proteins, lipoproteins and nucleic acids provide essential microorganism-specific gene products necessary to support microbial structure, function and viability, many of these same components are also shed from the outer cell wall of different Gram-negative bacterial species into surrounding biofluids which eventually enter the systemic circulation. Several of these microbial-derived secreted molecular species represent some of the most pro-inflammatory and noxious neurotoxins known. These neurotoxins disrupt cell-cell adhesion and easily translocate across aged or damaged plasma membranes and into the systemic circulation, brain, and CNS and PNS compartments. For example, microbial lipoprotein glycoconjugates such as Gram-negative bacteria-derived lipopolysaccharide (LPS), bacterial amyloids and more recently small non-coding RNA (sncRNA) microbial-derived neurotoxins have been found by many independent research groups to reside within the brain cells and CNS tissues of aged patients affected with Alzheimer's disease (AD). This 'Commentary' will highlight the most recent findings on these microbial-derived secreted toxins, their neurotropic properties and the potential contribution of these neurotoxic and pro-inflammatory microbial exudates to age-related inflammatory neurodegeneration, with specific reference to the human GI-tract abundant Gram-negative anaerobe Bacteroides fragilis and to AD wherever possible.
Collapse
Affiliation(s)
- Yuhai Zhao
- Department of Anatomy and Cell Biology, Louisiana State University, New Orleans, USA,LSU Neuroscience Center, Louisiana State University Health Science Center, New Orleans, USA
| | - Vivian Jaber
- LSU Neuroscience Center, Louisiana State University Health Science Center, New Orleans, USA
| | - Walter J. Lukiw
- LSU Neuroscience Center, Louisiana State University Health Science Center, New Orleans, USA,Department of Ophthalmology, LSU Neuroscience Center Louisiana State University Health Science Center, New Orleans, USA,Department Neurology, LSU Neuroscience Center Louisiana State University Health Science Center, New Orleans, USA
| |
Collapse
|
29
|
Lukiw WJ, Arceneaux L, Li W, Bond T, Zhao Y. Gastrointestinal (GI)-Tract Microbiome Derived Neurotoxins and their Potential Contribution to Inflammatory Neurodegeneration in Alzheimer's Disease (AD). JOURNAL OF ALZHEIMER'S DISEASE & PARKINSONISM 2021; 11:525. [PMID: 34457996 PMCID: PMC8395586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The human gastrointestinal (GI)-tract microbiome is a rich, complex and dynamic source of microorganisms that possess a staggering diversity and complexity. Importantly there is a significant variability in microbial complexity even amongst healthy individuals-this has made it difficult to link specific microbial abundance patterns with age-related neurological disease. GI-tract commensal microorganisms are generally beneficial to human metabolism and immunity, however enterotoxigenic forms of microbes possess significant potential to secrete what are amongst the most neurotoxic and pro-inflammatory biopolymers known. These include toxic glycolipids such as lipopolysaccharide (LPS), enterotoxins, microbial-derived amyloids and small non-coding RNA. One major microbial species of the GI-tract microbiome, about ~100-fold more abundant than Escherichia coli in deep GI-tract regions is Bacteroides fragilis, an anaerobic, rod-shaped Gram-negative bacterium. B. fragilis can secrete: (i) a particularly potent, pro-inflammatory and unique LPS subtype (BF-LPS); and (ii) a zinc-metalloproteinase known as B. fragilis-toxin (BFT) or fragilysin. Ongoing studies indicate that BF-LPS and/or BFT disrupt paracellular-and transcellular-barriers by cleavage of intercellular-proteins resulting in 'leaky' barriers. These barriers: (i) become defective and more penetrable with aging and disease; and (ii) permit entry of microbiome-derived neurotoxins into the systemic-circulation from which they next transit the blood-brain barrier and gain access to the CNS. Here LPS accumulates and significantly alters homeostatic patterns of gene expression. The affinity of LPS for neuronal nuclei is significantly enhanced in the presence of amyloid beta 42 (Aβ42) peptides. Recent research on the appearance of the brain thanatomicrobiome at the time of death and the increasing likelihood of a complex brain microbiome are reviewed and discussed. This paper will also highlight some recent advances in this extraordinary research area that links the pro-inflammatory exudates of the GI-tract microbiome with innate-immune disturbances and inflammatory-signaling within the CNS with reference to Alzheimer's disease (AD) wherever possible.
Collapse
Affiliation(s)
- Walter J. Lukiw
- LSU Neuroscience Center, Louisiana State University Health
Sciences Center, New Orleans, LA, United States,Department of Ophthalmology, LSU Health Sciences Center,
New Orleans, LA, United States,Department of Neurology, Louisiana State University Health
Sciences Center, New Orleans, LA, United States,Corresponding author: Dr. Walter J. Lukiw, LSU
Neuroscience Center, Louisiana State University Health Sciences Center, New
Orleans, LA, United States,
| | - Lisa Arceneaux
- LSU Neuroscience Center, Louisiana State University Health
Sciences Center, New Orleans, LA, United States
| | - Wenhong Li
- LSU Neuroscience Center, Louisiana State University Health
Sciences Center, New Orleans, LA, United States,Department of Pharmacology, School of Pharmacy, Jiangxi
University of Traditional Chinese Medicine (TCM), Nanchang, China
| | - Taylor Bond
- LSU Neuroscience Center, Louisiana State University Health
Sciences Center, New Orleans, LA, United States
| | - Yuhai Zhao
- LSU Neuroscience Center, Louisiana State University Health
Sciences Center, New Orleans, LA, United States,Department of Anatomy and Cell Biology, Louisiana State
University, New Orleans, LA, United States
| |
Collapse
|
30
|
Rodrigues GZP, Finkler M, Garcia ALH, Gehlen G. Evaluation of transgenerational effects caused by metals as environmental pollutants in Daphnia magna. ENVIRONMENTAL MONITORING AND ASSESSMENT 2020; 192:755. [PMID: 33170361 DOI: 10.1007/s10661-020-08713-4] [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: 07/03/2020] [Accepted: 10/26/2020] [Indexed: 06/11/2023]
Abstract
The present study aimed to evaluate the acute and chronic toxicity of environmentally relevant concentrations of metals (Mn, Al, Fe, and Pb) in Daphnia magna and the generational transposition of reproductive and morphological damages. The effective concentration for 10% of the organisms from each metal was obtained by the acute toxicity test (96 hours); then, another five concentrations lower than this one were defined for the chronic experimentation (21 days), in which the number of neonates generated by each individual was checked daily. At the end of the exposition, the lengths and number of morphological damages were recorded in each adult daphnid. During this, the molt generated on the 14th and 21st days were collected and cultivated for posterior evaluation of the same parameters. Alterations in the reproductive performance were observed in the organisms exposed to manganese and aluminum (4.0 and 0.5 mg L-1, respectively). Organisms exposed to aluminum (0.05 mg L-1) and iron (0.27 mg L-1) showed a reduction in body length. It is also noteworthy that the molt of these adults and their respective offspring also presented reproductive alterations, especially the molt from the 14th day of lead exposure (0.02 mg L-1) and the 21st day of manganese exposure (4.0 mg L-1). Such effects allow us to conclude that environments polluted by metals can reduce the ability of the species to maintain themselves in the ecosystem. In addition, there is a need to increase the control and monitoring of metals, such as aluminum, which present risks even in low concentrations.
Collapse
Affiliation(s)
| | | | - Ana Letícia Hilario Garcia
- Post Graduation Program in Cellular and Molecular Biology Applied to Health, ULBRA - Lutheran University of Brazil, Farroupilha Avenue, 8001, Canoas, Brazil
| | - Günther Gehlen
- Post Graduation Program in Environmental Quality, Feevale University, ERS-239, 2755, Novo Hamburgo, 93525-075, Brazil
| |
Collapse
|
31
|
Aplysin Retards Pancreatic Necrosis and Inflammatory Responses in NOD Mice by Stabilizing Intestinal Barriers and Regulating Gut Microbial Composition. Mediators Inflamm 2020; 2020:1280130. [PMID: 32801992 PMCID: PMC7416259 DOI: 10.1155/2020/1280130] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 06/14/2020] [Accepted: 06/19/2020] [Indexed: 01/26/2023] Open
Abstract
Aplysin is a brominated sesquiterpene with an isoprene skeleton and has biological activities. The purpose of this study is to investigate the inhibitory effect of aplysin on spontaneous pancreatic necrosis in nonobese diabetic (NOD) mice and its potential mechanisms. Results showed that NOD mice at 12 weeks of age showed obvious spontaneous pancreatic necrosis, damaged tight junctions of intestinal epithelia, and widened gaps in tight and adherens junctions. Aplysin intervention was able to alleviate spontaneous pancreatic necrosis in NOD mice, accompanied with decreased serum endotoxin levels and downregulated expressions of Toll-like receptor 4 and its related molecules MyD88, TRAF-6, NF-κB p65, TRIF, TRAM, and IRF-3, as well as protein levels of interleukin-1β and interferon-β in pancreatic tissues. In addition, we observed obvious improvements of intestinal mucosal barrier function and changes of gut microbiota in the relative abundance at the phylum level and the genus level in aplysin-treated mice compared with control mice. Together, these data suggested that aplysin could retard spontaneous pancreatic necrosis and inflammatory responses in NOD mice through the stabilization of intestinal barriers and regulation of gut microbial composition.
Collapse
|
32
|
Lukiw WJ. Human gastrointestinal (GI) tract microbiome-derived pro-inflammatory neurotoxins from Bacteroides fragilis: Effects of low fiber diets and environmental and lifestyle factors. INTEGRATIVE FOOD, NUTRITION AND METABOLISM 2020; 7:277. [PMID: 33381303 PMCID: PMC7771874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Affiliation(s)
- Walter J Lukiw
- LSU Neuroscience Center, Louisiana State University Health Sciences Center, New Orleans LA 70112 USA
- Department of Ophthalmology, Louisiana State University Health Sciences Center, New Orleans LA 70112 USA
- Department of Neurology, Louisiana State University Health Sciences Center, New Orleans LA 70112 USA
| |
Collapse
|