51
|
da Silva Rodrigues F, Jantsch J, de Farias Fraga G, Luiza de Camargo Milczarski V, Silva Dias V, Scheid C, de Oliveira Merib J, Giovernardi M, Padilha Guedes R. Cannabidiol improves maternal obesity-induced behavioral, neuroinflammatory and neurochemical dysfunctions in the juvenile offspring. Brain Behav Immun 2024; 119:301-316. [PMID: 38608740 DOI: 10.1016/j.bbi.2024.04.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 03/25/2024] [Accepted: 04/09/2024] [Indexed: 04/14/2024] Open
Abstract
Maternal obesity is associated with an increased risk of psychiatric disorders such as anxiety, depression, schizophrenia and autism spectrum disorder in the offspring. While numerous studies focus on preventive measures targeting the mothers, only a limited number provide practical approaches for addressing the damages once they are already established. We have recently demonstrated the interplay between maternal obesity and treatment with cannabidiol (CBD) on hypothalamic inflammation and metabolic disturbances, however, little is known about this relationship on behavioral manifestations and neurochemical imbalances in other brain regions. Therefore, here we tested whether CBD treatment could mitigate anxiety-like and social behavioral alterations, as well as neurochemical disruptions in both male and female offspring of obese dams. Female Wistar rats were fed a cafeteria diet for 12 weeks prior to mating, and during gestation and lactation. Offspring received CBD (50 mg/kg) from weaning for 3 weeks. Behavioral tests assessed anxiety-like manifestations and social behavior, while neuroinflammatory and neurochemical markers were evaluated in the prefrontal cortex (PFC) and hippocampus. CBD treatment attenuated maternal obesity-induced anxiety-like and social behavioral alterations, followed by rescuing effects on imbalanced neurotransmitter and endocannabinoid concentrations and altered expression of glial markers, CB1, oxytocin and dopamine receptors, with important differences between sexes. Overall, the findings of this study provide insight into the signaling pathways for the therapeutic benefits of CBD on neuroinflammation and neurochemical imbalances caused by perinatal maternal obesity in the PFC and the hippocampus, which translates into the behavioral manifestations, highlighting the sexual dimorphism encompassing both the transgenerational effect of obesity and the endocannabinoid system.
Collapse
Affiliation(s)
- Fernanda da Silva Rodrigues
- Graduate Program in Biosciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), 90050-170, Porto Alegre, Rio Grande do Sul, Brazil
| | - Jeferson Jantsch
- Graduate Program in Biosciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), 90050-170, Porto Alegre, Rio Grande do Sul, Brazil
| | - Gabriel de Farias Fraga
- Graduate Program in Biosciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), 90050-170, Porto Alegre, Rio Grande do Sul, Brazil
| | - Vitória Luiza de Camargo Milczarski
- Undergraduate Program in Biomedical Sciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), 90050-170, Porto Alegre, Rio Grande do Sul, Brazil
| | - Victor Silva Dias
- Undergraduate Program in Biomedical Sciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), 90050-170, Porto Alegre, Rio Grande do Sul, Brazil
| | - Camila Scheid
- Graduate Program in Biosciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), 90050-170, Porto Alegre, Rio Grande do Sul, Brazil
| | - Josias de Oliveira Merib
- Graduate Program in Biosciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), 90050-170, Porto Alegre, Rio Grande do Sul, Brazil
| | - Marcia Giovernardi
- Graduate Program in Biosciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), 90050-170, Porto Alegre, Rio Grande do Sul, Brazil; Graduate Program in Health Sciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, 90050-170 Rio Grande do Sul, Brazil
| | - Renata Padilha Guedes
- Graduate Program in Biosciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), 90050-170, Porto Alegre, Rio Grande do Sul, Brazil; Graduate Program in Health Sciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, 90050-170 Rio Grande do Sul, Brazil.
| |
Collapse
|
52
|
Ren Q, Zhu X, Pan J, Li K, Zhou Y, Lyu Y, Xie Q, Xu Y. A combination of phospholipids and long chain polyunsaturated fatty acids supports neurodevelopmental outcomes in infants: a randomized, double-blind, controlled clinical trial. Front Nutr 2024; 11:1358651. [PMID: 38938667 PMCID: PMC11208465 DOI: 10.3389/fnut.2024.1358651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 06/03/2024] [Indexed: 06/29/2024] Open
Abstract
Phospholipids (PLs) and long-chain polyunsaturated fatty acids (LCPUFAs) are naturally present in breast milk and play important roles in promoting the growth of the infant. Several studies have investigated the effects of the combination of PLs and LCPUFAs on neurodevelopment. However, data on the effectiveness of infant formula containing both PLs and LCPUFAs on the neurodevelopment of infants is still scarce. This randomized, double-blind, controlled clinical study was designed to evaluate the effect of an infant formula enriched with PLs and LCPUFAs on growth parameters and neurodevelopmental outcomes in term infants up to 365 days of age. Infants were enrolled within 30 days of birth who were then randomly assigned to either a control group (n = 150) or an investigational group (n = 150). Both groups consist of cow's milk-based formula which were generally identical in terms of composition, except that the investigational formula was additionally supplemented with PLs and LCPUFAs. The infants were followed for the first year of life. Breastfed infants were the reference (n = 150). Bayley Scales of Infant Development [3rd edition (Bayley-III)], Carey Toddler Temperament Scales (TTS), MacArthur-Bates Communicative Development Inventories (CDI), Single Object Attention and Free Play Tasks were used to evaluate neurodevelopmental outcomes of infant at 365 days of age. In addition, Ages and Stages Questionnaires (ASQ) were also conducted at 120, 180, and 275 days of age. Compared to breastfeeding, both infant formulas were well-tolerated and provided adequate growth, with no adverse events being reported throughout the study. Infants of the investigational group showed higher mean scores in Bayley-III cognitive performance (104.3 vs. 99.0, p < 0.05), language (106.9 vs. 104.5, p < 0.05), and motor skills (109.2 vs. 103.9, p < 0.05) compared the control group. Similar results were being reported for other developmental scales including TTS and ASQ. Notably, the test scores of infants fed the investigational formula were similar to those who were breastfed. Our results indicate that PL and LCPUFA supplementation may be beneficial for neurodevelopment of infants throughout the first year of life. Further studies are needed to investigation long-term effects PL and LCPUFA on neurodevelopment in early life.
Collapse
Affiliation(s)
- Qiqi Ren
- Heilongjiang Feihe Dairy Co., Ltd., Beijing, China
- PKUHSC-China Feihe Joint Research Institute of Nutrition and Healthy Lifespan Development, Beijing, China
| | - Xiaoyu Zhu
- PKUHSC-China Feihe Joint Research Institute of Nutrition and Healthy Lifespan Development, Beijing, China
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing, China
- Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Peking University, Beijing, China
| | - Jiancun Pan
- Heilongjiang Feihe Dairy Co., Ltd., Beijing, China
- PKUHSC-China Feihe Joint Research Institute of Nutrition and Healthy Lifespan Development, Beijing, China
| | - Kaifeng Li
- Heilongjiang Feihe Dairy Co., Ltd., Beijing, China
- PKUHSC-China Feihe Joint Research Institute of Nutrition and Healthy Lifespan Development, Beijing, China
| | - Yalin Zhou
- PKUHSC-China Feihe Joint Research Institute of Nutrition and Healthy Lifespan Development, Beijing, China
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing, China
- Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Peking University, Beijing, China
| | - Ying Lyu
- PKUHSC-China Feihe Joint Research Institute of Nutrition and Healthy Lifespan Development, Beijing, China
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing, China
- Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Peking University, Beijing, China
| | - Qinggang Xie
- Heilongjiang Feihe Dairy Co., Ltd., Beijing, China
- PKUHSC-China Feihe Joint Research Institute of Nutrition and Healthy Lifespan Development, Beijing, China
| | - Yajun Xu
- PKUHSC-China Feihe Joint Research Institute of Nutrition and Healthy Lifespan Development, Beijing, China
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing, China
- Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Peking University, Beijing, China
| |
Collapse
|
53
|
Liu A, Li Y, Li L, Chen K, Tan M, Zou F, Zhang X, Meng X. Bile acid metabolism is altered in learning and memory impairment induced by chronic lead exposure. JOURNAL OF HAZARDOUS MATERIALS 2024; 471:134360. [PMID: 38663295 DOI: 10.1016/j.jhazmat.2024.134360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 04/07/2024] [Accepted: 04/18/2024] [Indexed: 05/12/2024]
Abstract
Lead is a neurotoxic contaminant that exists widely in the environment. Although lead neurotoxicity has been found to be tightly linked to gut microbiota disturbance, the effect of host metabolic disorders caused by gut microbiota disturbance on lead neurotoxicity has not been investigated. In this work, the results of new object recognition tests and Morris water maze tests showed that chronic low-dose lead exposure caused learning and memory dysfunction in mice. The results of 16 S rRNA sequencing of cecal contents and fecal microbiota transplantation showed that the neurotoxicity of lead could be transmitted through gut microbiota. The results of untargeted metabolomics and bile acid targeted metabolism analysis showed that the serum bile acid metabolism profile of lead-exposed mice was significantly changed. In addition, supplementation with TUDCA or INT-777 significantly alleviated chronic lead exposure-induced learning and memory impairment, primarily through inhibition of the NLRP3 inflammasome in the hippocampus to relieve neuroinflammation. In conclusion, our findings suggested that dysregulation of host bile acid metabolism may be one of the mechanisms of lead-induced neurotoxicity, and supplementation of specific bile acids may be a possible therapeutic strategy for lead-induced neurotoxicity.
Collapse
Affiliation(s)
- Anfei Liu
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou 510515, China
| | - Yunting Li
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou 510515, China
| | - Lifan Li
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou 510515, China
| | - Kaiju Chen
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou 510515, China
| | - Meitao Tan
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou 510515, China
| | - Fei Zou
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou 510515, China
| | - Xingmei Zhang
- Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Xiaojing Meng
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou 510515, China.
| |
Collapse
|
54
|
Romero‐Marco P, Chicharro C, Verde Z, Miguel‐Tobal F, Fernández‐Araque A. Effect on blood lipids and body composition of a high-fat (MUFA) and high-fiber diet: A case-control study. Food Sci Nutr 2024; 12:3863-3871. [PMID: 38873480 PMCID: PMC11167160 DOI: 10.1002/fsn3.4042] [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: 05/16/2023] [Revised: 01/29/2024] [Accepted: 02/07/2024] [Indexed: 06/15/2024] Open
Abstract
Metabolic Syndrome (MetS) is a constellation of risk factors including abdominal obesity, high triglycerides, low HDL cholesterol (HDL-C), elevated blood pressure, and elevated fasting glucose. In Spain, according to WHO criteria, the MetS prevalence is shown to be 32% in men and 29% in women. The role of dietary habits is one of the main therapeutic strategies for the management of MetS but the most effective dietary pattern has not been established yet. This study aimed to analyze the effect of on body composition, serum lipids, and MetS components of a high-MUFA and high-fiber diet (HMFD). A case-control study was performed considering 40 cohabiting women. Participants were randomly assigned to HMFD group or high mono-unsaturated diet (HMD) group to receive one of the two proposed dietary interventions. All data (serum lipids, blood pressure, height, weight, body composition, and waist circumference) were collected fasting at baseline, 55, 98, and 132 days. The HMFD group showed higher decrease in waist circumference than in the HMD group. LDL-C dropped in both groups. Triglycerides in the HMFD group dropped during the intervention, but once the intervention was over, they returned to baseline values. The mean systolic blood pressure was lower in HMFD group. A HMFD from a weekly consumption of processed meat (Torrezno de Soria) deeply fried in extra virgin olive oil in combination with vegetables logged in a Mediterranean diet can improve MetS risk factors in healthy overweight women.
Collapse
Affiliation(s)
- Patricia Romero‐Marco
- Department of Nursing, Faculty of Health SciencesUniversity of ValladolidSoriaSpain
- Pharmacogenetics, Cancer Genetics, Genetic Polymorphisms and Pharmacoepidemiology, Center for Drug Safety Studies, Department of Nursing, Health Sciences, Molecular Genetics of Disease – IBGMUniversity of ValladolidSoriaSpain
| | - Celia Chicharro
- Department of Legal Medicine, Psychiatry and Pathology, Biopathology‐Toxicology Laboratory, Faculty of MedicineUniversity Complutense of MadridMadridSpain
- Centro de Estudios Gregorio MarañónFundación Ortega‐MarañónMadridSpain
| | - Zoraida Verde
- Pharmacogenetics, Cancer Genetics, Genetic Polymorphisms and Pharmacoepidemiology, Center for Drug Safety Studies, Department of Nursing, Health Sciences, Molecular Genetics of Disease – IBGMUniversity of ValladolidSoriaSpain
- Centro de Estudios Gregorio MarañónFundación Ortega‐MarañónMadridSpain
- Department of Biochemistry and Molecular Biology, Faculty of Health SciencesUniversity of ValladolidSoriaSpain
| | - Francisco Miguel‐Tobal
- Department of Radiology, Rehabilitation and Physiotherapy; School of Medicine of Physical Education and Sport; Faculty of MedicineUniversity Complutense of MadridMadridSpain
| | - Ana Fernández‐Araque
- Department of Nursing, Faculty of Health SciencesUniversity of ValladolidSoriaSpain
- Pharmacogenetics, Cancer Genetics, Genetic Polymorphisms and Pharmacoepidemiology, Center for Drug Safety Studies, Department of Nursing, Health Sciences, Molecular Genetics of Disease – IBGMUniversity of ValladolidSoriaSpain
| |
Collapse
|
55
|
Xu Y, Qiu Z, Gu C, Yu S, Wang S, Li C, Yao X, Li W. Propionate alleviates itch in murine models of atopic dermatitis by modulating sensory TRP channels of dorsal root ganglion. Allergy 2024; 79:1271-1290. [PMID: 38164798 DOI: 10.1111/all.15998] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 12/07/2023] [Accepted: 12/10/2023] [Indexed: 01/03/2024]
Abstract
BACKGROUND Itch is the most common symptom of atopic dermatitis (AD) and significantly decreases the quality of life. Skin microbiome is involved in AD pathogenesis, whereas its role in the regulation of itch remains elusive. In this study, we aimed to investigate the effects of skin microbial metabolite propionate on acute and chronic pruritus and to explore the mechanism. METHODS Using various mouse models of itch, the roles of propionate were explored by behavioral tests and histopathology/immunofluorescent analysis. Primary-cultured dorsal root ganglion neurons and HEK293 cells expressing recombinant human TRP channels were utilized for in vitro calcium imaging/in vivo miniature two-photon imaging in combination with electrophysiology and molecular docking approaches for investigation of the mechanism. RESULTS Propionate significantly alleviated itch and alloknesis in various mouse models of pruritus and AD and decreased the density of intraepidermal nerve fibers. Propionate reduced the responsiveness of dorsal root ganglion neurons to pruritogens in vitro, attenuated the hyper-excitability in sensory neurons in MC903-induced AD model, and inhibited capsaicin-evoked hTRPV1 currents (IC50 = 20.08 ± 1.11 μM) via interacting with the vanilloid binding site. Propionate also decreased the secretion of calcitonin gene-related peptide by nerves in MC903-induced AD mouse model, which further attenuated itch and skin inflammation. CONCLUSION Our study revealed a protective effect of propionate against persistent itch through direct modulation of sensory TRP channels and neuropeptide production in neurons. Regulation of itch via the skin microbiome might be a novel strategy for the treatment of AD.
Collapse
Affiliation(s)
- Yao Xu
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai Institute of Dermatology, Shanghai, China
| | - Zhuoqiong Qiu
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai Institute of Dermatology, Shanghai, China
| | - Chaoying Gu
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai Institute of Dermatology, Shanghai, China
| | - Su Yu
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai Institute of Dermatology, Shanghai, China
| | - Shangshang Wang
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai Institute of Dermatology, Shanghai, China
| | - Changlin Li
- Guangdong Institute of Intelligence Science and Technology, Zhuhai, China
| | - Xu Yao
- Department of Allergy and Rheumatology, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Hospital for skin diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Wei Li
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai Institute of Dermatology, Shanghai, China
| |
Collapse
|
56
|
Babaei M, Machle CJ, Mokhtari P, Ottino González J, Schmidt KA, Alderete TL, Adise S, Peterson BS, Goran MI. Pre-pregnancy maternal obesity and infant neurodevelopmental outcomes in Latino infants. Obesity (Silver Spring) 2024; 32:979-988. [PMID: 38600046 DOI: 10.1002/oby.24010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 01/17/2024] [Accepted: 01/19/2024] [Indexed: 04/12/2024]
Abstract
OBJECTIVE This study explores the impact of maternal pre-pregnancy BMI on infant neurodevelopment at 24 months in low-income Latino families. It also investigates whether infant diet mediates this relationship. METHODS Latino mother-infant pairs (n = 163) were enrolled at 1 month post partum and were followed for 2 years, with assessments at 6-month intervals. Maternal pre-pregnancy anthropometrics were self-reported at baseline, and child neurodevelopment was assessed at 24 months using the Bayley Scales of Infant Development. Diet quality of infants was measured using the Healthy Eating Index (HEI)-2015 and HEI-Toddlers-2020 scores at multiple time points. Mediation and regression models that adjust for maternal factors were used to examine the associations. RESULTS Pre-pregnancy BMI showed significant negative associations with child cognitive scores (β = -0.1, 95% CI: -0.2 to -0.06, p < 0.001) and language scores (β = -0.1, 95% CI: -0.2 to -0.03, p = 0.01) at 24 months. Infant HEI-2015 scores at 24 months partly mediated these associations, explaining 23% and 30% of the total effect on cognitive and language subscales, respectively. No specific dietary components in infants mediated the relationship, except for the total HEI-2015 score. CONCLUSIONS Managing maternal obesity pre-pregnancy is crucial for improving infant neurodevelopmental outcomes, especially in low-income Latino families. Promoting healthy weight and enhancing infant diet quality can enhance neurodevelopment in these populations.
Collapse
Affiliation(s)
- Mahsa Babaei
- Department of Pediatrics, Division of Endocrinology, Diabetes, and Metabolism, Children's Hospital Los Angeles, Los Angeles, California, USA
| | - Christopher J Machle
- Department of Pediatrics, Division of Endocrinology, Diabetes, and Metabolism, Children's Hospital Los Angeles, Los Angeles, California, USA
- Department of Psychology, University of Oregon, Eugene, Oregon, USA
| | - Pari Mokhtari
- Department of Pediatrics, Division of Endocrinology, Diabetes, and Metabolism, Children's Hospital Los Angeles, Los Angeles, California, USA
| | - Jonatan Ottino González
- Department of Pediatrics, Division of Endocrinology, Diabetes, and Metabolism, Children's Hospital Los Angeles, Los Angeles, California, USA
| | - Kelsey A Schmidt
- Department of Pediatrics, Division of Endocrinology, Diabetes, and Metabolism, Children's Hospital Los Angeles, Los Angeles, California, USA
| | - Tanya L Alderete
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, USA
| | - Shana Adise
- Department of Pediatrics, Division of Endocrinology, Diabetes, and Metabolism, Children's Hospital Los Angeles, Los Angeles, California, USA
| | - Bradley S Peterson
- Department of Pediatrics, Division of Endocrinology, Diabetes, and Metabolism, Children's Hospital Los Angeles, Los Angeles, California, USA
| | - Michael I Goran
- Department of Pediatrics, Division of Endocrinology, Diabetes, and Metabolism, Children's Hospital Los Angeles, Los Angeles, California, USA
| |
Collapse
|
57
|
Chen B, de Launoit E, Renier N, Schneeberger M. Maternal nutritional programming shapes the cerebral landscape. Trends Endocrinol Metab 2024; 35:367-370. [PMID: 37949733 DOI: 10.1016/j.tem.2023.10.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Revised: 10/20/2023] [Accepted: 10/23/2023] [Indexed: 11/12/2023]
Abstract
The escalating prevalence of maternal obesity raises concerns about its influence on offspring health. Exposure to obesogenic environments during early development leads to persistent alterations in brain function contributing to neurological disorders. Nutritional programming emerges as a promising avenue to counteract the deleterious effects of maternal obesity on offspring neurodevelopment.
Collapse
Affiliation(s)
- Bandy Chen
- Laboratory of Neurovascular Control of Homeostasis, Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT, USA
| | - Elisa de Launoit
- Laboratoire de Plasticité Structurale Sorbonne Université, ICM Paris Brain Institute, INSERM U1127, CNRS UMR7225, 75013 Paris, France
| | - Nicolas Renier
- Laboratoire de Plasticité Structurale Sorbonne Université, ICM Paris Brain Institute, INSERM U1127, CNRS UMR7225, 75013 Paris, France
| | - Marc Schneeberger
- Laboratory of Neurovascular Control of Homeostasis, Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT, USA; Wu Tsai Institute for Mind and Brain, Yale University, New Haven, CT, USA.
| |
Collapse
|
58
|
Ren SJ, Feng JT, Xiang T, Liao CL, Zhou YP, Xuan RR. Expression and clinical significance of short-chain fatty acids in patients with intrahepatic cholestasis of pregnancy. World J Hepatol 2024; 16:601-611. [PMID: 38689740 PMCID: PMC11056904 DOI: 10.4254/wjh.v16.i4.601] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 02/04/2024] [Accepted: 03/07/2024] [Indexed: 04/24/2024] Open
Abstract
BACKGROUND Intrahepatic cholestasis of pregnancy (ICP) is a pregnancy-specific liver condition that typically arises in the middle and late stages of pregnancy. Short-chain fatty acids (SCFAs), prominent metabolites of the gut microbiota, have significant connections with various pregnancy complications, and some SCFAs hold potential for treating such complications. However, the metabolic profile of SCFAs in patients with ICP remains unclear. AIM To investigate the metabolic profiles and differences in SCFAs present in the maternal and cord blood of patients with ICP and determine the clinical significance of these findings. METHODS Maternal serum and cord blood samples were collected from both patients with ICP (ICP group) and normal pregnant women (NP group). Targeted metabolomics was used to assess the SCFA levels in these samples. RESULTS Significant differences in maternal SCFAs were observed between the ICP and NP groups. Most SCFAs exhibited a consistent declining trend in cord blood samples from the ICP group, mirroring the pattern seen in maternal serum. Correlation analysis revealed a positive correlation between maternal serum SCFAs and cord blood SCFAs [r (Pearson) = 0.88, P = 7.93e-95]. In both maternal serum and cord blood, acetic and caproic acids were identified as key metabolites contributing to the differences in SCFAs between the two groups (variable importance for the projection > 1). Receiver operating characteristic analysis demonstrated that multiple SCFAs in maternal blood have excellent diagnostic capabilities for ICP, with caproic acid exhibiting the highest diagnostic efficacy (area under the curve = 0.97). CONCLUSION Compared with the NP group, significant alterations were observed in the SCFAs of maternal serum and cord blood in the ICP group, although they displayed distinct patterns of change. Furthermore, the SCFA levels in maternal serum and cord blood were significantly positively correlated. Notably, certain maternal serum SCFAs, specifically caproic and acetic acids, demonstrated excellent diagnostic efficiency for ICP.
Collapse
Affiliation(s)
- Shuai-Jun Ren
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Ningbo University, Ningbo 315100, Zhejiang Province, China
| | - Jia-Ting Feng
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Ningbo University, Ningbo 315100, Zhejiang Province, China
- Health Science Center, Ningbo University, Ningbo 315211, Zhejiang Province, China
| | - Ting Xiang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Ningbo University, Ningbo 315100, Zhejiang Province, China
- Health Science Center, Ningbo University, Ningbo 315211, Zhejiang Province, China
| | - Cai-Lian Liao
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Ningbo University, Ningbo 315100, Zhejiang Province, China
- Health Science Center, Ningbo University, Ningbo 315211, Zhejiang Province, China
| | - Yu-Ping Zhou
- Department of Gastroenterology, The First Affiliated Hospital of Ningbo University, Ningbo 315211, Zhejiang Province, China
- Institute of Digestive Disease, Ningbo University, Ningbo 315020, Zhejiang Province, China
| | - Rong-Rong Xuan
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Ningbo University, Ningbo 315100, Zhejiang Province, China.
| |
Collapse
|
59
|
Wang Y, Xie D, Ma S, Shao N, Zhang X, Wang X. Exploring the common mechanism of vascular dementia and inflammatory bowel disease: a bioinformatics-based study. Front Immunol 2024; 15:1347415. [PMID: 38736878 PMCID: PMC11084673 DOI: 10.3389/fimmu.2024.1347415] [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: 11/30/2023] [Accepted: 04/15/2024] [Indexed: 05/14/2024] Open
Abstract
Objective Emerging evidence has shown that gut diseases can regulate the development and function of the immune, metabolic, and nervous systems through dynamic bidirectional communication on the brain-gut axis. However, the specific mechanism of intestinal diseases and vascular dementia (VD) remains unclear. We designed this study especially, to further clarify the connection between VD and inflammatory bowel disease (IBD) from bioinformatics analyses. Methods We downloaded Gene expression profiles for VD (GSE122063) and IBD (GSE47908, GSE179285) from the Gene Expression Omnibus (GEO) database. Then individual Gene Set Enrichment Analysis (GSEA) was used to confirm the connection between the two diseases respectively. The common differentially expressed genes (coDEGs) were identified, and the STRING database together with Cytoscape software were used to construct protein-protein interaction (PPI) network and core functional modules. We identified the hub genes by using the Cytohubba plugin. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis were applied to identify pathways of coDEGs and hub genes. Subsequently, receiver operating characteristic (ROC) analysis was used to identify the diagnostic ability of these hub genes, and a training dataset was used to verify the expression levels of the hub genes. An alternative single-sample gene set enrichment (ssGSEA) algorithm was used to analyze immune cell infiltration between coDEGs and immune cells. Finally, the correlation between hub genes and immune cells was analyzed. Results We screened 167 coDEGs. The main articles of coDEGs enrichment analysis focused on immune function. 8 shared hub genes were identified, including PTPRC, ITGB2, CYBB, IL1B, TLR2, CASP1, IL10RA, and BTK. The functional categories of hub genes enrichment analysis were mainly involved in the regulation of immune function and neuroinflammatory response. Compared to the healthy controls, abnormal infiltration of immune cells was found in VD and IBD. We also found the correlation between 8 shared hub genes and immune cells. Conclusions This study suggests that IBD may be a new risk factor for VD. The 8 hub genes may predict the IBD complicated with VD. Immune-related coDEGS may be related to their association, which requires further research to prove.
Collapse
Affiliation(s)
- Yujiao Wang
- Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Daojun Xie
- Encephalopathy Center, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Shijia Ma
- Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Nan Shao
- Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Xiaoyan Zhang
- Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Xie Wang
- Anhui University of Chinese Medicine, Hefei, Anhui, China
| |
Collapse
|
60
|
Saadh MJ, Ahmed HM, Alani ZK, Al Zuhairi RAH, Almarhoon ZM, Ahmad H, Ubaid M, Alwan NH. The Role of Gut-derived Short-Chain Fatty Acids in Multiple Sclerosis. Neuromolecular Med 2024; 26:14. [PMID: 38630350 DOI: 10.1007/s12017-024-08783-4] [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: 02/04/2024] [Accepted: 03/08/2024] [Indexed: 04/19/2024]
Abstract
Multiple sclerosis (MS) is a chronic condition affecting the central nervous system (CNS), where the interplay of genetic and environmental factors influences its pathophysiology, triggering immune responses and instigating inflammation. Contemporary research has been notably dedicated to investigating the contributions of gut microbiota and their metabolites in modulating inflammatory reactions within the CNS. Recent recognition of the gut microbiome and dietary patterns as environmental elements impacting MS development emphasizes the potential influence of small, ubiquitous molecules from microbiota, such as short-chain fatty acids (SCFAs). These molecules may serve as vital molecular signals or metabolic substances regulating host cellular metabolism in the intricate interplay between microbiota and the host. A current emphasis lies on optimizing the health-promoting attributes of colonic bacteria to mitigate urinary tract issues through dietary management. This review aims to spotlight recent investigations on the impact of SCFAs on immune cells pivotal in MS, the involvement of gut microbiota and SCFAs in MS development, and the considerable influence of probiotics on gastrointestinal disruptions in MS. Comprehending the gut-CNS connection holds promise for the development of innovative therapeutic approaches, particularly probiotic-based supplements, for managing MS.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman, 11831, Jordan
| | - Hani Moslem Ahmed
- Department of Dental Industry Techniques, Al-Noor University College, Nineveh, Iraq
| | - Zaid Khalid Alani
- College of Health and Medical Technical, Al-Bayan University, Baghdad, Iraq
| | | | - Zainab M Almarhoon
- Department of Chemistry, College of Science, King Saud University, 11451, Riyadh, Saudi Arabia
| | - Hijaz Ahmad
- Section of Mathematics, International Telematic University Uninettuno, Corso Vittorio Emanuele II, 39, 00186, Rome, Italy.
- Center for Applied Mathematics and Bioinformatics, Gulf University for Science and Technology, Mubarak Al-Abdullah, Kuwait.
- Department of Computer Science and Mathematics, Lebanese American University, Beirut, Lebanon.
| | - Mohammed Ubaid
- Medical Technical College, Al-Farahidi University, Baghdad, Iraq
| | | |
Collapse
|
61
|
Lu X, Shi Z, Jiang L, Zhang S. Maternal gut microbiota in the health of mothers and offspring: from the perspective of immunology. Front Immunol 2024; 15:1362784. [PMID: 38545107 PMCID: PMC10965710 DOI: 10.3389/fimmu.2024.1362784] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 02/28/2024] [Indexed: 04/17/2024] Open
Abstract
Due to the physiological alteration during pregnancy, maternal gut microbiota changes following the metabolic processes. Recent studies have revealed that maternal gut microbiota is closely associated with the immune microenvironment in utero during pregnancy and plays a vital role in specific pregnancy complications, including preeclampsia, gestational diabetes, preterm birth and recurrent miscarriages. Some other evidence has also shown that aberrant maternal gut microbiota increases the risk of various diseases in the offspring, such as allergic and neurodevelopmental disorders, through the immune alignment between mother and fetus and the possible intrauterine microbiota. Probiotics and the high-fiber diet are effective inventions to prevent mothers and fetuses from diseases. In this review, we summarize the role of maternal gut microbiota in the development of pregnancy complications and the health condition of future generations from the perspective of immunology, which may provide new therapeutic strategies for the health management of mothers and offspring.
Collapse
Affiliation(s)
- Xiaowen Lu
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Obstetrics and Gynecology, Key Laboratory of Reproductive Dysfunction, Management of Zhejiang Province, Hangzhou, China
| | - Zhan Shi
- Department of Obstetrics and Gynecology, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu, China
| | - Lingling Jiang
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Obstetrics and Gynecology, Key Laboratory of Reproductive Dysfunction, Management of Zhejiang Province, Hangzhou, China
| | - Songying Zhang
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Obstetrics and Gynecology, Key Laboratory of Reproductive Dysfunction, Management of Zhejiang Province, Hangzhou, China
| |
Collapse
|
62
|
Lv JJ, Li XY, Wang JB, Yang XT, Yin MY, Yang CH. Association of dietary live microbe intake with various cognitive domains in US adults aged 60 years or older. Sci Rep 2024; 14:5714. [PMID: 38459061 PMCID: PMC10923796 DOI: 10.1038/s41598-024-51520-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 01/06/2024] [Indexed: 03/10/2024] Open
Abstract
The purpose of this study was to explore whether dietary live microbe intake is associated with various cognitive domains using data from the National Health and Nutrition Examination Survey (NHANES) from 2011 to 2014. And the specific relationship between low, medium and high dietary live microbe intake groups and cognitive ability of the elderly. Dietary live microbe intake was calculated from 24-h diet recall interviews. Cognitive function was assessed using the number symbol substitution test (DSST, which measures processing speed), the animal fluency test (AFT, which measures executive function), the Alzheimer's Registry sub-test (CERAD, which measures memory), and the Composite Z-score, which adds the Z-values of individual tests. Multiple linear regression models and restricted cubic bar graphs were used to investigate the relationship between live microbe intake and cognitive performance. A total of 2,450 participants aged 60 or older were included. Live microbe intake was positively correlated with cognitive ability on the whole. Specifically, when the intake of low, medium and high live microbe was > 2640 g, > 39 g and > 0 g respectively, the CERAD, DSST, AFT and compositive-Z score of the subjects increased with the increase of microbial intake (P < 0.05). In American adults age 60 or older, higher intakes of live microbes were associated with better cognitive performance, especially after a certain amount was reached.
Collapse
Affiliation(s)
- Jia-Jie Lv
- Department of Vascular Surgery, Shanghai Putuo People's Hospital, School of Medicine, Tongji University, No.1291 Jiangning Road, Huangpu District, Shanghai, 200011, China
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, No.639 Zhizaoju Road, Huangpu District, Shanghai, 200011, China
| | - Xin-Yu Li
- Department of Vascular Surgery, Shanghai Putuo People's Hospital, School of Medicine, Tongji University, No.1291 Jiangning Road, Huangpu District, Shanghai, 200011, China
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No.639 Zhizaoju Road, Huangpu District, Shanghai, 200011, China
| | - Jing-Bing Wang
- Department of Interventional Therapy, Multidisciplinary Team of Vascular Anomalies, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University, No.639 Zhizaoju Road, Huangpu District, Shanghai, 200011, China
| | - Xi-Tao Yang
- Department of Interventional Therapy, Multidisciplinary Team of Vascular Anomalies, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University, No.639 Zhizaoju Road, Huangpu District, Shanghai, 200011, China
| | - Min-Yi Yin
- Department of Vascular Surgery, Shanghai Putuo People's Hospital, School of Medicine, Tongji University, No.1291 Jiangning Road, Huangpu District, Shanghai, 200011, China
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, No.639 Zhizaoju Road, Huangpu District, Shanghai, 200011, China
| | - Cheng-Hao Yang
- Department of Vascular Surgery, Shanghai Putuo People's Hospital, School of Medicine, Tongji University, No.1291 Jiangning Road, Huangpu District, Shanghai, 200011, China.
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, No.639 Zhizaoju Road, Huangpu District, Shanghai, 200011, China.
| |
Collapse
|
63
|
Mei X, Li Y, Zhang X, Zhai X, Yang Y, Li Z, Li L. Maternal Phlorizin Intake Protects Offspring from Maternal Obesity-Induced Metabolic Disorders in Mice via Targeting Gut Microbiota to Activate the SCFA-GPR43 Pathway. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:4703-4725. [PMID: 38349207 DOI: 10.1021/acs.jafc.3c06370] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/07/2024]
Abstract
Maternal obesity increases the risk of obesity and metabolic disorders (MDs) in offspring, which can be mediated by the gut microbiota. Phlorizin (PHZ) can improve gut dysbiosis and positively affect host health; however, its transgenerational metabolic benefits remain largely unclear. This study aimed to investigate the potential of maternal PHZ intake in attenuating the adverse impacts of a maternal high-fat diet on obesity-related MDs in dams and offspring. The results showed that maternal PHZ reduced HFD-induced body weight gain and fat accumulation and improved glucose intolerance and abnormal lipid profiles in both dams and offspring. PHZ improved gut dysbiosis by promoting expansion of SCFA-producing bacteria, Akkermansia and Blautia, while inhibiting LPS-producing and pro-inflammatory bacteria, resulting in significantly increased fecal SCFAs, especially butyric acid, and reduced serum lipopolysaccharide levels and intestinal inflammation. PHZ also promoted intestinal GLP-1/2 secretion and intestinal development and enhanced gut barrier function by activating G protein-coupled receptor 43 (GPR43) in the offspring. Antibiotic-treated mice receiving FMT from PHZ-regulated offspring could attenuate MDs induced by receiving FMT from HFD offspring through the gut microbiota to activate the GPR43 pathway. It can be regarded as a promising functional food ingredient for preventing intergenerational transmission of MDs and breaking the obesity cycle.
Collapse
Affiliation(s)
- Xueran Mei
- Department of Obstetrics, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen 518020, China
- Post-Doctoral Scientific Research Station of Clinical Medicine, Jinan University, Guangzhou 510632, China
- Department of Obstetrics, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai 201204, China
| | - Yi Li
- Graduate School of Biomedical Engineering, Faculty of Engineering, University of New South Wales, Sydney 2052, Australia
- ARC Centre of Excellence for Nanoscale Biophotonics, University of New South Wales, Sydney 2052, Australia
| | - Xiaoyu Zhang
- College of Life Sciences, Sichuan Normal University, Chengdu 610101, China
| | - Xiwen Zhai
- Graduate School of Biomedical Engineering, Faculty of Engineering, University of New South Wales, Sydney 2052, Australia
- ARC Centre of Excellence for Nanoscale Biophotonics, University of New South Wales, Sydney 2052, Australia
| | - Yi Yang
- Department of Obstetrics, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen 518020, China
- Post-Doctoral Scientific Research Station of Clinical Medicine, Jinan University, Guangzhou 510632, China
| | - Zhengjuan Li
- Department of Obstetrics, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen 518020, China
- Post-Doctoral Scientific Research Station of Clinical Medicine, Jinan University, Guangzhou 510632, China
- Department of Obstetrics, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai 201204, China
| | - Liping Li
- Department of Obstetrics, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen 518020, China
- Post-Doctoral Scientific Research Station of Clinical Medicine, Jinan University, Guangzhou 510632, China
- Department of Obstetrics, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai 201204, China
| |
Collapse
|
64
|
Yin X, Liu W, Feng H, Huang J, Wang Q, Zhang Q, He J, Wang R. Bifidobacterium animalis subsp. lactis A6 attenuates hippocampal damage and memory impairments in an ADHD rat model. Food Funct 2024; 15:2668-2678. [PMID: 38374797 DOI: 10.1039/d3fo04665f] [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: 02/21/2024]
Abstract
Attention deficit hyperactivity disorder (ADHD) is commonly accompanied by learning and memory deficits. This study aimed to demonstrate the effects of probiotic Bifidobacterium animalis subsp. lactis A6 (BAA6) on behaviour and memory function in spontaneously hypertensive rats (SHRs). The results showed that BAA6 treatment ameliorated spatial working memory deficits and inhibited hippocampal neuron loss in SHRs. The levels of neurotransmitters such as acetylcholine, dopamine, and norepinephrine, and the brain derived neurotrophic factor increased and that of glutamate decreased in the brain tissue of SHRs after BAA6 administration. Moreover, BAA6 reduced the levels of pro-inflammatory cytokines TNF-α and IL-1β, and increased the levels of anti-inflammatory IL-10 and antioxidant glutathione in SHRs. 16S rRNA high-throughput sequencing showed that BAA6 treatment changed the gut microbiota composition. BAA6 promoted beneficial Lactobacillus, Romboutsia, Blautia, and Turicibacter, and decreased the enrichment of bacterial genera such as Dietzia, Sporosarcina, Brevibacterium, NK4A214_group, Atopostipes, and Facklamia negatively associated with neurotransmitter release and anti-inflammatory effects in SHRs. Together, these results suggested that BAA6 improved memory function by ameliorating hippocampal damage, abnormal neurotransmitter release and cerebral inflammation by reshaping the gut microbiota in SHRs. This study provides a scientific basis for the development and application of BAA6 as a promising dietary intervention to reduce the risk of ADHD.
Collapse
Affiliation(s)
- Xindi Yin
- Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing 100190, China.
| | - Weichen Liu
- Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing 100190, China.
- National Center of Technology Innovation for Dairy, Hohhot 010000, China
| | - Haihong Feng
- Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing 100190, China.
- Hebei Engineering Research Center of Animal Product, Langfang 065200, China
| | - Jiaqiang Huang
- Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing 100190, China.
| | - Qi Wang
- Xinjiang Golden Camel Investment Co., Ltd., Wulumuqi 830039, China
| | - Qi Zhang
- Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing 100190, China.
| | - Jingjing He
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China.
| | - Ran Wang
- Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing 100190, China.
| |
Collapse
|
65
|
Loh JS, Mak WQ, Tan LKS, Ng CX, Chan HH, Yeow SH, Foo JB, Ong YS, How CW, Khaw KY. Microbiota-gut-brain axis and its therapeutic applications in neurodegenerative diseases. Signal Transduct Target Ther 2024; 9:37. [PMID: 38360862 PMCID: PMC10869798 DOI: 10.1038/s41392-024-01743-1] [Citation(s) in RCA: 215] [Impact Index Per Article: 215.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 01/02/2024] [Accepted: 01/14/2024] [Indexed: 02/17/2024] Open
Abstract
The human gastrointestinal tract is populated with a diverse microbial community. The vast genetic and metabolic potential of the gut microbiome underpins its ubiquity in nearly every aspect of human biology, including health maintenance, development, aging, and disease. The advent of new sequencing technologies and culture-independent methods has allowed researchers to move beyond correlative studies toward mechanistic explorations to shed light on microbiome-host interactions. Evidence has unveiled the bidirectional communication between the gut microbiome and the central nervous system, referred to as the "microbiota-gut-brain axis". The microbiota-gut-brain axis represents an important regulator of glial functions, making it an actionable target to ameliorate the development and progression of neurodegenerative diseases. In this review, we discuss the mechanisms of the microbiota-gut-brain axis in neurodegenerative diseases. As the gut microbiome provides essential cues to microglia, astrocytes, and oligodendrocytes, we examine the communications between gut microbiota and these glial cells during healthy states and neurodegenerative diseases. Subsequently, we discuss the mechanisms of the microbiota-gut-brain axis in neurodegenerative diseases using a metabolite-centric approach, while also examining the role of gut microbiota-related neurotransmitters and gut hormones. Next, we examine the potential of targeting the intestinal barrier, blood-brain barrier, meninges, and peripheral immune system to counteract glial dysfunction in neurodegeneration. Finally, we conclude by assessing the pre-clinical and clinical evidence of probiotics, prebiotics, and fecal microbiota transplantation in neurodegenerative diseases. A thorough comprehension of the microbiota-gut-brain axis will foster the development of effective therapeutic interventions for the management of neurodegenerative diseases.
Collapse
Affiliation(s)
- Jian Sheng Loh
- School of Pharmacy, Monash University Malaysia, Jalan Lagoon Selatan, 47500, Bandar Sunway, Selangor, Malaysia
| | - Wen Qi Mak
- School of Pharmacy, Monash University Malaysia, Jalan Lagoon Selatan, 47500, Bandar Sunway, Selangor, Malaysia
| | - Li Kar Stella Tan
- School of Pharmacy, Faculty of Health & Medical Sciences, Taylor's University, 1, Jalan Taylors, Subang Jaya, 47500, Selangor, Malaysia
- Digital Health & Medical Advancements, Taylor's University, 1, Jalan Taylors, Subang Jaya, 47500, Selangor, Malaysia
| | - Chu Xin Ng
- School of Biosciences, Faculty of Health & Medical Sciences, Taylor's University, 1, Jalan Taylors, Subang Jaya, 47500, Selangor, Malaysia
| | - Hong Hao Chan
- School of Pharmacy, Monash University Malaysia, Jalan Lagoon Selatan, 47500, Bandar Sunway, Selangor, Malaysia
| | - Shiau Hueh Yeow
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London, WC1N 1AX, UK
| | - Jhi Biau Foo
- School of Pharmacy, Faculty of Health & Medical Sciences, Taylor's University, 1, Jalan Taylors, Subang Jaya, 47500, Selangor, Malaysia
- Digital Health & Medical Advancements, Taylor's University, 1, Jalan Taylors, Subang Jaya, 47500, Selangor, Malaysia
| | - Yong Sze Ong
- School of Pharmacy, Monash University Malaysia, Jalan Lagoon Selatan, 47500, Bandar Sunway, Selangor, Malaysia
| | - Chee Wun How
- School of Pharmacy, Monash University Malaysia, Jalan Lagoon Selatan, 47500, Bandar Sunway, Selangor, Malaysia.
| | - Kooi Yeong Khaw
- School of Pharmacy, Monash University Malaysia, Jalan Lagoon Selatan, 47500, Bandar Sunway, Selangor, Malaysia.
| |
Collapse
|
66
|
Lu D, Liu Y, Kang L, Zhang X, Hu J, Ye H, Huang B, Wu Y, Zhao J, Dai Z, Wang J, Han D. Maternal fiber-rich diet promotes early-life intestinal development in offspring through milk-derived extracellular vesicles carrying miR-146a-5p. J Nanobiotechnology 2024; 22:65. [PMID: 38365722 PMCID: PMC10870446 DOI: 10.1186/s12951-024-02344-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 02/12/2024] [Indexed: 02/18/2024] Open
Abstract
BACKGROUNDS The intestinal development in early life is profoundly influenced by multiple biological components of breast milk, in which milk-derived extracellular vesicles (mEVs) contain a large amount of vertically transmitted signal from the mother. However, little is known about how maternal fiber-rich diet regulates offspring intestinal development by influencing the mEVs. RESULTS In this study, we found that maternal resistant starch (RS) consumption during late gestation and lactation improved the growth and intestinal health of offspring. The mEVs in breast milk are the primary factor driving these beneficial effects, especially enhancing intestinal cell proliferation and migration. To be specific, administration of mEVs after maternal RS intake enhanced intestinal cell proliferation and migration in vivo (performed in mice model and indicated by intestinal histological observation, EdU assay, and the quantification of cyclin proteins) and in vitro (indicated by CCK8, MTT, EdU, and wound healing experiments). Noteworthily, miR-146a-5p was found to be highly expressed in the mEVs from maternal RS group, which also promotes intestinal cell proliferation in cells and mice models. Mechanically, miR-146a-5p target to silence the expression of ubiquitin ligase 3 gene NEDD4L, thereby inhibiting DVL2 ubiquitination, activating the Wnt pathway, and promoting intestinal development. CONCLUSION These findings demonstrated the beneficial role of mEVs in the connection between maternal fiber rich diet and offspring intestinal growth. In addition, we identified a novel miRNA-146a-5p-NEDD4L-β-catenin/Wnt signaling axis in regulating early intestinal development. This work provided a new perspective for studying the influence of maternal diet on offspring development.
Collapse
Affiliation(s)
- Dongdong Lu
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
- Adaptation Physiology Group, Wageningen University & Research, Wageningen, 6700 AH, The Netherlands
| | - Yisi Liu
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Luyuan Kang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Xiangyu Zhang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Jie Hu
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Hao Ye
- Adaptation Physiology Group, Wageningen University & Research, Wageningen, 6700 AH, The Netherlands
| | - Bingxu Huang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Yujun Wu
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Jinbiao Zhao
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Zhaolai Dai
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Junjun Wang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Dandan Han
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
67
|
Zhang T, Cheng T, Geng S, Mao K, Li X, Gao J, Han J, Sang Y. Synbiotic Combination between Lactobacillus paracasei VL8 and Mannan-Oligosaccharide Repairs the Intestinal Barrier in the Dextran Sulfate Sodium-Induced Colitis Model by Regulating the Intestinal Stem Cell Niche. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:2214-2228. [PMID: 38237048 DOI: 10.1021/acs.jafc.3c08473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
Previously, Lactobacillus paracasei VL8, a lactobacillus strain isolated from the traditional Finnish fermented dairy product Viili, demonstrated immunomodulatory and antibacterial effects. The prebiotic mannan-oligosaccharide (MOS) further promoted its antibacterial activity and growth performance, holding promise for maintaining intestinal health. However, this has not been verified in vivo. In this study, we elucidated the process by which L. paracasei VL8 and its synbiotc combination (SYN) with MOS repair the intestinal barrier function in dextran sodium sulfate (DSS)-induced colitis mice. SYN surpasses VL8 or MOS alone in restoring goblet cells and improving the tight junction structure. Omics analysis on gut microbiota reveals SYN's ability to restore Lactobacillus spp. abundance and promote tryptophan metabolism. SYN intervention also inhibits the DSS-induced hyperactivation of the Wnt/β-catenin pathway. Tryptophan metabolites from Lactobacillus induce intestinal organoid differentiation. Co-housing experiments confirm microbiota transferability, replicating intestinal barrier repair. In conclusion, our study highlights the potential therapeutic efficacy of the synbiotic combination of Lactobacillus paracasei VL8 and MOS in restoring the damaged intestinal barrier and offers new insights into the complex crosstalk between the gut microbiota and intestinal stem cells.
Collapse
Affiliation(s)
- Tuo Zhang
- Department of Food Science and Technology, Hebei Agricultural University, Baoding, Hebei CN 071000, China
| | - Tiantian Cheng
- Department of Food Science and Technology, Hebei Agricultural University, Baoding, Hebei CN 071000, China
| | - Shuo Geng
- Department of Food Science and Technology, Hebei Agricultural University, Baoding, Hebei CN 071000, China
| | - Kemin Mao
- Department of Food Science and Technology, Hebei Agricultural University, Baoding, Hebei CN 071000, China
| | - Xiyu Li
- Department of Food Science and Technology, Hebei Agricultural University, Baoding, Hebei CN 071000, China
| | - Jie Gao
- Department of Food Science and Technology, Hebei Agricultural University, Baoding, Hebei CN 071000, China
| | - Jun Han
- Department of Food Science and Technology, Hebei Agricultural University, Baoding, Hebei CN 071000, China
| | - Yaxin Sang
- Department of Food Science and Technology, Hebei Agricultural University, Baoding, Hebei CN 071000, China
| |
Collapse
|
68
|
Kadyan S, Park G, Hochuli N, Miller K, Wang B, Nagpal R. Resistant starches from dietary pulses improve neurocognitive health via gut-microbiome-brain axis in aged mice. Front Nutr 2024; 11:1322201. [PMID: 38352704 PMCID: PMC10864001 DOI: 10.3389/fnut.2024.1322201] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Accepted: 01/10/2024] [Indexed: 02/16/2024] Open
Abstract
Introduction Cognitive decline is a common consequence of aging. Dietary patterns that lack fibers and are high in saturated fats worsen cognitive impairment by triggering pro-inflammatory pathways and metabolic dysfunctions. Emerging evidence highlights the neurocognitive benefits of fiber-rich diets and the crucial role of gut-microbiome-brain signaling. However, the mechanisms of this diet-microbiome-brain regulation remain largely unclear. Methods Accordingly, we herein investigated the unexplored neuroprotective mechanisms of dietary pulses-derived resistant starch (RS) in improving aging-associated neurocognitive function in an aged (60-weeks old) murine model carrying a human microbiome. Results and discussion Following 20-weeks dietary regimen which included a western-style diet without (control; CTL) or with 5% w/w fortification with RS from pinto beans (PTB), black-eyed-peas (BEP), lentils (LEN), chickpeas (CKP), or inulin fiber (INU), we find that RS, particularly from LEN, ameliorate the cognitive impairments induced by western diet. Mechanistically, RS-mediated improvements in neurocognitive assessments are attributed to positive remodeling of the gut microbiome-metabolome arrays, which include increased short-chain fatty acids and reduced branched-chain amino acids levels. This microbiome-metabolite-brain signaling cascade represses neuroinflammation, cellular senescence, and serum leptin/insulin levels, while enhancing lipid metabolism through improved hepatic function. Altogether, the data demonstrate the prebiotic effects of RS in improving neurocognitive function via modulating the gut-brain axis.
Collapse
Affiliation(s)
- Saurabh Kadyan
- The Gut Biome Lab, Department of Health, Nutrition, and Food Sciences, College of Education, Health, and Human Sciences, Florida State University, Tallahassee, FL, United States
| | - Gwoncheol Park
- The Gut Biome Lab, Department of Health, Nutrition, and Food Sciences, College of Education, Health, and Human Sciences, Florida State University, Tallahassee, FL, United States
| | - Nathaniel Hochuli
- The Gut Biome Lab, Department of Health, Nutrition, and Food Sciences, College of Education, Health, and Human Sciences, Florida State University, Tallahassee, FL, United States
| | - Katelyn Miller
- The Gut Biome Lab, Department of Health, Nutrition, and Food Sciences, College of Education, Health, and Human Sciences, Florida State University, Tallahassee, FL, United States
| | - Bo Wang
- Department of Biomedical and Chemical Engineering and Sciences, Florida Institute of Technology, Melbourne, FL, United States
| | - Ravinder Nagpal
- The Gut Biome Lab, Department of Health, Nutrition, and Food Sciences, College of Education, Health, and Human Sciences, Florida State University, Tallahassee, FL, United States
| |
Collapse
|
69
|
Wang C, Chen W, Jiang Y, Xiao X, Zou Q, Liang J, Zhao Y, Wang Q, Yuan T, Guo R, Liu X, Liu Z. A synbiotic formulation of Lactobacillus reuteri and inulin alleviates ASD-like behaviors in a mouse model: the mediating role of the gut-brain axis. Food Funct 2024; 15:387-400. [PMID: 38099485 DOI: 10.1039/d3fo02663a] [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: 01/03/2024]
Abstract
Autism Spectrum Disorder (ASD), a complex neurodevelopmental disorder marked by social communication deficits and repetitive behaviors, may see symptom amelioration through gut microbiota modulation. This study investigates the effects of a synbiotic - specifically a probiotic amplified by prebiotic supplementation - on ASD-like mouse model's social deficiencies. This model was established via valproic acid injection into pregnant females. Post-weaning, male progeny received daily synbiotic treatment, a combination of Lactobacillus reuteri (L. reuteri) and inulin, for four weeks. Results indicated that the synbiotic rectified social impairments and attenuated inflammatory cytokine expressions in the brain. Moreover, synbiotic intervention protected gut barrier integrity and altered the gut microbiota composition, enhancing the butyrate-producing Bifidobacterium abundance. The synbiotic elevated metabolites such as butyrate and 3-hydroxybutyric acid (3-HB), alongside upregulated genes associated with 3-HB synthesis in the colon and liver, and brain receptors. Conclusively, the synbiotic combination of L. reuteri and inulin mitigated ASD-related social impairments, partially via their regulatory effect on the gut-brain axis.
Collapse
Affiliation(s)
- Chuanchuan Wang
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi 712100, China.
- Northwest A&F University Shenzhen Research Institute, Shenzhen, Guangdong, 518000, China
| | - Weixuan Chen
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi 712100, China.
| | - Yishan Jiang
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi 712100, China.
| | - Xiao Xiao
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi 712100, China.
| | - Qianhui Zou
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi 712100, China.
| | - Jiarui Liang
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi 712100, China.
| | - Yu Zhao
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi 712100, China.
| | - Qianxu Wang
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi 712100, China.
| | - Tian Yuan
- Northwest A&F University Shenzhen Research Institute, Shenzhen, Guangdong, 518000, China
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, College of Chemistry & Pharmacy, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Rui Guo
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi 712100, China.
- Northwest A&F University Shenzhen Research Institute, Shenzhen, Guangdong, 518000, China
| | - Xuebo Liu
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi 712100, China.
| | - Zhigang Liu
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi 712100, China.
- Northwest A&F University Shenzhen Research Institute, Shenzhen, Guangdong, 518000, China
| |
Collapse
|
70
|
Li X, Zheng P, Zou Y, Guan L, Li N, Liu J, Lu N, Zhu Y, He C. Dietary inulin ameliorates obesity-induced severe acute pancreatitis via gut-pancreas axis. Gut Microbes 2024; 16:2436949. [PMID: 39653685 PMCID: PMC11633219 DOI: 10.1080/19490976.2024.2436949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 11/04/2024] [Accepted: 11/26/2024] [Indexed: 12/13/2024] Open
Abstract
Obesity is a definitive factor of severity and mortality of acute pancreatitis (AP), and gut microbiota dysbiosis is involved in its pathogenesis. However, the effect of gut microbiota modulation by dietary components on high fat diet (HFD)-induced severe AP remains unclear. Here, we found that the inulin, a soluble dietary fiber, mitigated pancreatic injury and systematic inflammation in mice fed HFD, which was dependent on gut microbiota as this protective effect was attenuated in germ-free mice. Inulin treatment suppressed the overgrowth of pathogenic bacteria Escherichia Shigella, Enterococcus, Klebsiella, while increased the abundance of probiotics Akkermansia. Fecal microbiota transplantation from inulin-treated mice to recipient mice reduced pancreatic damage and remodeled intestinal homeostasis. Additionally, inulin increased fecal short chain fatty acids (SCFAs), strengthened gut barrier and restored Paneth cells. The beneficial effect of inulin on improving pancreatic damage and leaky gut was diminished after the suppression of SCFAs. Notably, SCFAs administration, especially butyrate, to HFD mice blocked pancreatic and intestinal injury with the inhibition of histone deacetylase 3 (HDAC3), and pharmacological HDAC3 inhibition mimicked the ameliorative effect of SCFAs. Mechanically, butyrate modulated macrophage M1/M2 polarization balance by suppressing HDAC3 and subsequent acetylation of histone H3K27. Collectively, our data offer new insights into the gut microbiota-pancreas axis that may be leveraged to augment the potential supplementation of prebiotic inulin in the management of obesity associated severe AP.
Collapse
Affiliation(s)
- Xin Li
- Department of Gastroenterology, Jiangxi Provincial Key Laboratory of Digestive Diseases, Jiangxi Clinical Research Center for Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Pan Zheng
- Department of Gastroenterology, Jiangxi Provincial Key Laboratory of Digestive Diseases, Jiangxi Clinical Research Center for Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Yaoyu Zou
- Department of Gastroenterology, Jiangxi Provincial Key Laboratory of Digestive Diseases, Jiangxi Clinical Research Center for Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Langyi Guan
- Department of Gastroenterology, Jiangxi Provincial Key Laboratory of Digestive Diseases, Jiangxi Clinical Research Center for Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Nianshuang Li
- Department of Gastroenterology, Jiangxi Provincial Key Laboratory of Digestive Diseases, Jiangxi Clinical Research Center for Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Jianping Liu
- Department of Gastroenterology, Jiangxi Provincial Key Laboratory of Digestive Diseases, Jiangxi Clinical Research Center for Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Nonghua Lu
- Department of Gastroenterology, Jiangxi Provincial Key Laboratory of Digestive Diseases, Jiangxi Clinical Research Center for Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Yin Zhu
- Department of Gastroenterology, Jiangxi Provincial Key Laboratory of Digestive Diseases, Jiangxi Clinical Research Center for Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Jiangxi Medicine Academy of Nutrition and Health Management, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Cong He
- Department of Gastroenterology, Jiangxi Provincial Key Laboratory of Digestive Diseases, Jiangxi Clinical Research Center for Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
71
|
Li L, Liu T, Shi Y. Treatment of preterm brain injury via gut-microbiota-metabolite-brain axis. CNS Neurosci Ther 2024; 30:e14556. [PMID: 38108213 PMCID: PMC10805406 DOI: 10.1111/cns.14556] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 11/06/2023] [Accepted: 11/23/2023] [Indexed: 12/19/2023] Open
Abstract
BACKGROUND Brain injury in preterm infants potentially disrupts critical structural and functional connective networks in the brain. It is a major cause of neurological sequelae and developmental deficits in preterm infants. Interesting findings suggest that the gut microbiota (GM) and their metabolites contribute to the programming of the central nervous system (CNS) during developmental stages and may exert structural and functional effects throughout the lifespan. AIM To summarize the existing knowledge of the potential mechanisms related to immune, endocrine, neural, and blood-brain barrier (BBB) mediated by GM and its metabolites in neural development and function. METHODS We review the recent literature and included 150 articles to summarize the mechanisms through which GM and their metabolites work on the nervous system. Potential health benefits and challenges of relevant treatments are also discussed. RESULTS This review discusses the direct and indirect ways through which the GM may act on the nervous system. Treatment of preterm brain injury with GM or related derivatives, including probiotics, prebiotics, synbiotics, dietary interventions, and fecal transplants are also included. CONCLUSION This review summarizes mechanisms underlying microbiota-gut-brain axis and novel therapeutic opportunities for neurological sequelae in preterm infants. Optimizing the initial colonization and microbiota development in preterm infants may represent a novel therapy to promote brain development and reduce long-term sequelae.
Collapse
Affiliation(s)
- Ling Li
- Department of PediatricsShengjing Hospital of China Medical UniversityShenyangChina
| | - Tianjing Liu
- Department of PediatricsShengjing Hospital of China Medical UniversityShenyangChina
| | - Yongyan Shi
- Department of PediatricsShengjing Hospital of China Medical UniversityShenyangChina
| |
Collapse
|
72
|
Zhang H, Li C, Han L, Xiao Y, Bian J, Liu C, Gong L, Liu Z, Wang M. MUP1 mediates urolithin A alleviation of chronic alcohol-related liver disease via gut-microbiota-liver axis. Gut Microbes 2024; 16:2367342. [PMID: 38889450 PMCID: PMC11188796 DOI: 10.1080/19490976.2024.2367342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 06/07/2024] [Indexed: 06/20/2024] Open
Abstract
Alcohol-related liver disease (ALD) is recognized as a global health crisis, contributing to approximately 20% of liver cancer-associated fatalities. Dysbiosis of the gut microbiome is associated with the development of ALD, with the gut microbial metabolite urolithin A (UA) exhibiting a potential for alleviating liver symptoms. However, the protective efficacy of UA against ALD and its underlying mechanism mediated by microbiota remain elusive. In this study, we provide evidence demonstrating that UA effectively ameliorates alcohol-induced metabolic disorders and hepatic endoplasmic reticulum (ER) stress through a specific gut-microbiota-liver axis mediated by major urinary protein 1 (MUP1). Moreover, UA exhibited the potential to restore alcohol-induced dysbiosis of the intestinal microbiota by enriching the abundance of Bacteroides sartorii (B. sartorii), Parabacteroides distasonis (P. distasonis), and Akkermansia muciniphila (A. muciniphila), along with their derived metabolite propionic acid. Partial attenuation of the hepatoprotective effects exerted by UA was observed upon depletion of gut microbiota using antibiotics. Subsequently, a fecal microbiota transplantation (FMT) experiment was conducted to evaluate the microbiota-dependent effects of UA in ALD. FMT derived from mice treated with UA exhibited comparable efficacy to direct UA treatment, as it effectively attenuated ER stress through modulation of MUP1. It was noteworthy that strong associations were observed among the hepatic MUP1, gut microbiome, and metabolome profiles affected by UA. Intriguingly, oral administration of UA-enriched B. sartorii, P. distasonis, and A. muciniphila can enhance propionic acid production to effectively suppress ER stress via MUP1, mimicking UA treatment. Collectively, these findings elucidate the causal mechanism that UA alleviated ALD through the gut-microbiota-liver axis. This unique mechanism sheds light on developing novel microbiome-targeted therapeutic strategies against ALD.
Collapse
Affiliation(s)
- Hongbo Zhang
- College of Food Science and Engineering, Northwest A&F University, Yang ling, Shaanxi, China
| | - Chaoyue Li
- College of Food Science and Engineering, Northwest A&F University, Yang ling, Shaanxi, China
| | - Lin Han
- College of Food Science and Engineering, Northwest A&F University, Yang ling, Shaanxi, China
| | - Yao Xiao
- College of Food Science and Engineering, Northwest A&F University, Yang ling, Shaanxi, China
| | - Ji Bian
- Kolling Institute, Sydney Medical School, Royal North Shore Hospital, University of Sydney, Sydney, Australia
| | - Chao Liu
- Key Laboratory of Novel Food Resources Processing, Ministry of Agriculture and Rural Affairs/Institute of Agro-Food Science and Technology, Shandong Academy of Agricultural Sciences, Jinan, P.R. China
| | - Lan Gong
- UNSW Microbiome Research Centre, St George and Sutherland Clinical Campus, University of New South Wales, Sydney, Australia
| | - Zhigang Liu
- College of Food Science and Engineering, Northwest A&F University, Yang ling, Shaanxi, China
| | - Min Wang
- College of Food Science and Engineering, Northwest A&F University, Yang ling, Shaanxi, China
| |
Collapse
|
73
|
Chen M, Zhao Y, Ji H, Li L, Liu H, Wang S, Zhang D, Yin J, Wang J, Zhang X. Chenodeoxycholic Acid Improves Embryo Implantation and Metabolic Health through Modulating Gut Microbiota-Host Metabolites Interaction during Early Pregnancy. Antioxidants (Basel) 2023; 13:8. [PMID: 38275628 PMCID: PMC10812749 DOI: 10.3390/antiox13010008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 12/14/2023] [Accepted: 12/15/2023] [Indexed: 01/27/2024] Open
Abstract
Fetus loss in early pregnancy is of major concern to both humans and animals, and this issue is largely influenced by embryo implantation. Chenodeoxycholic acid (CDCA), a primary bile acid, contributes to metabolic improvements and protects against intrahepatic cholestasis of pregnancy. However, the effect of CDCA on embryo implantation during early pregnancy has not been investigated. The present study demonstrated that CDCA administration during early pregnancy improved embryo implantation in sows and rats, thereby improving the pregnancy outcomes of sows. CDCA significantly reduced inflammation, oxidative stress, and insulin resistance. The metabolomics analysis indicated significant differences in the fecal metabolome, especially regarding the level of secondary bile acids, between the control and CDCA-treated sows. CDCA also influenced the serum metabolite profiles in sows, and the serum L-Histidine level was significantly correlated with the abundance of 19 differential fecal metabolites. Importantly, L-Histidine administration improved embryo implantation and metabolic health in rats during early pregnancy. Moreover, CDCA administration during early pregnancy also led to long-term metabolic improvements in sows. Our data indicated that CDCA improved embryo implantation by alleviating inflammation and oxidative stress, improving insulin sensitivity, and modulating the interaction between the gut microbiota and host metabolites. Therefore, CDCA intervention is a potential therapeutic strategy regarding embryo loss during pregnancy.
Collapse
Affiliation(s)
- Meixia Chen
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing 100097, China; (M.C.); (H.J.); (L.L.); (H.L.); (S.W.); (D.Z.)
| | - Ying Zhao
- Key Laboratory of Feed Biotechnology of the Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China;
| | - Haifeng Ji
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing 100097, China; (M.C.); (H.J.); (L.L.); (H.L.); (S.W.); (D.Z.)
| | - Lu Li
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing 100097, China; (M.C.); (H.J.); (L.L.); (H.L.); (S.W.); (D.Z.)
- College of Life Science and Food Engineering, Hebei University of Engineering, Handan 056038, China
| | - Hui Liu
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing 100097, China; (M.C.); (H.J.); (L.L.); (H.L.); (S.W.); (D.Z.)
| | - Sixin Wang
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing 100097, China; (M.C.); (H.J.); (L.L.); (H.L.); (S.W.); (D.Z.)
| | - Dongyan Zhang
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing 100097, China; (M.C.); (H.J.); (L.L.); (H.L.); (S.W.); (D.Z.)
| | - Jingdong Yin
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China;
| | - Jing Wang
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing 100097, China; (M.C.); (H.J.); (L.L.); (H.L.); (S.W.); (D.Z.)
| | - Xin Zhang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China;
| |
Collapse
|
74
|
Nohesara S, Abdolmaleky HM, Zhou JR, Thiagalingam S. Microbiota-Induced Epigenetic Alterations in Depressive Disorders Are Targets for Nutritional and Probiotic Therapies. Genes (Basel) 2023; 14:2217. [PMID: 38137038 PMCID: PMC10742434 DOI: 10.3390/genes14122217] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 12/08/2023] [Accepted: 12/11/2023] [Indexed: 12/24/2023] Open
Abstract
Major depressive disorder (MDD) is a complex disorder and a leading cause of disability in 280 million people worldwide. Many environmental factors, such as microbes, drugs, and diet, are involved in the pathogenesis of depressive disorders. However, the underlying mechanisms of depression are complex and include the interaction of genetics with epigenetics and the host immune system. Modifications of the gut microbiome and its metabolites influence stress-related responses and social behavior in patients with depressive disorders by modulating the maturation of immune cells and neurogenesis in the brain mediated by epigenetic modifications. Here, we discuss the potential roles of a leaky gut in the development of depressive disorders via changes in gut microbiota-derived metabolites with epigenetic effects. Next, we will deliberate how altering the gut microbiome composition contributes to the development of depressive disorders via epigenetic alterations. In particular, we focus on how microbiota-derived metabolites such as butyrate as an epigenetic modifier, probiotics, maternal diet, polyphenols, drugs (e.g., antipsychotics, antidepressants, and antibiotics), and fecal microbiota transplantation could positively alleviate depressive-like behaviors by modulating the epigenetic landscape. Finally, we will discuss challenges associated with recent therapeutic approaches for depressive disorders via microbiome-related epigenetic shifts, as well as opportunities to tackle such problems.
Collapse
Affiliation(s)
- Shabnam Nohesara
- Department of Medicine (Biomedical Genetics), Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA;
| | - Hamid Mostafavi Abdolmaleky
- Nutrition/Metabolism Laboratory, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boson, MA 02215, USA;
| | - Jin-Rong Zhou
- Nutrition/Metabolism Laboratory, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boson, MA 02215, USA;
| | - Sam Thiagalingam
- Department of Medicine (Biomedical Genetics), Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA;
- Department of Pathology & Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
| |
Collapse
|
75
|
Xu H, Yi X, Cui Z, Li H, Zhu L, Zhang L, Chen J, Fan X, Zhou P, Li MJ, Yu Y, Liu Q, Huang D, Yao Z, Zhou J. Maternal antibiotic exposure enhances ILC2 activation in neonates via downregulation of IFN1 signaling. Nat Commun 2023; 14:8332. [PMID: 38097561 PMCID: PMC10721923 DOI: 10.1038/s41467-023-43903-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 11/23/2023] [Indexed: 12/17/2023] Open
Abstract
Microbiota have an important function in shaping and priming neonatal immunity, although the cellular and molecular mechanisms underlying these effects remain obscure. Here we report that prenatal antibiotic exposure causes significant elevation of group 2 innate lymphoid cells (ILC2s) in neonatal lungs, in both cell numbers and functionality. Downregulation of type 1 interferon signaling in ILC2s due to diminished production of microbiota-derived butyrate represents the underlying mechanism. Mice lacking butyrate receptor GPR41 (Gpr41-/-) or type 1 interferon receptor IFNAR1 (Ifnar1-/-) recapitulate the phenotype of neonatal ILC2s upon maternal antibiotic exposure. Furthermore, prenatal antibiotic exposure induces epigenetic changes in ILC2s and has a long-lasting deteriorative effect on allergic airway inflammation in adult offspring. Prenatal supplementation of butyrate ameliorates airway inflammation in adult mice born to antibiotic-exposed dams. These observations demonstrate an essential role for the microbiota in the control of type 2 innate immunity at the neonatal stage, which suggests a therapeutic window for treating asthma in early life.
Collapse
Affiliation(s)
- Haixu Xu
- Department of Immunology, Tianjin Institute of Immunology, Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, State Key Laboratory of Experimental Hematology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Xianfu Yi
- Department of Bioinformatics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Zhaohai Cui
- Department of Immunology, Tianjin Institute of Immunology, Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, State Key Laboratory of Experimental Hematology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Hui Li
- Department of Immunology, Tianjin Institute of Immunology, Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, State Key Laboratory of Experimental Hematology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Lin Zhu
- Department of Immunology, Tianjin Institute of Immunology, Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, State Key Laboratory of Experimental Hematology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Lijuan Zhang
- Department of Immunology, Tianjin Institute of Immunology, Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, State Key Laboratory of Experimental Hematology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - JiaLe Chen
- Department of Immunology, Tianjin Institute of Immunology, Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, State Key Laboratory of Experimental Hematology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Xutong Fan
- Department of Bioinformatics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Pan Zhou
- Department of Immunology, Tianjin Institute of Immunology, Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, State Key Laboratory of Experimental Hematology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Mulin Jun Li
- Department of Bioinformatics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Ying Yu
- Department of Pharmacology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Qiang Liu
- Department of Neurology, Institute of Neuroimmunology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Dandan Huang
- Department of Pharmacology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China.
| | - Zhi Yao
- Department of Immunology, Tianjin Institute of Immunology, Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, State Key Laboratory of Experimental Hematology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China.
| | - Jie Zhou
- Department of Immunology, Tianjin Institute of Immunology, Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, State Key Laboratory of Experimental Hematology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China.
- Department of Neonatology, Guangzhou Key Laboratory of Neonatal Intestinal Diseases, the Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China.
| |
Collapse
|
76
|
Yan S, Yu L, Tian F, Zhao J, Chen W, Chen H, Zhai Q. Ligilactobacillus salivarius CCFM 1266 modulates gut microbiota and GPR109a-mediated immune suppression to attenuate immune checkpoint blockade-induced colitis. Food Funct 2023; 14:10549-10563. [PMID: 37953676 DOI: 10.1039/d3fo03867j] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2023]
Abstract
The wide application of immune checkpoint blockade (ICB) therapy is impeded by the development of ICB-induced colitis, a condition intricately linked to alterations in the gut microbiota. In our previous study, Ligilactobacillus salivarius CCFM 1266 and Bacteroides fragilis HCK-B3 exhibited anti-inflammatory properties. In this research, treatment with both L. salivarius CCFM 1266 and B. fragilis HCK-B3 significantly ameliorated body weight loss and colonic inflammation in murine colitis models induced by intravenous ipilimumab injection, with L. salivarius CCFM 1266 demonstrating superior effectiveness. This amelioration was characterized by an augmented ratio of Treg cells and M2 macrophages, a diminishment in pro-inflammatory cytokines (IL-1β, TNF-α, IFN-γ, IL-23), and an elevation in the anti-inflammatory cytokine IL-10. The ingestion of L. salivarius CCFM 1266 exerted a discernible influence on the composition of the gut microbiota. Untargeted metabolomics revealed an increase in colonic nicotinic acid levels following the administration of L. salivarius CCFM 1266, potentially initiating the activation of the colonic GPR109a pathway. This mechanism likely serves as the fundamental basis for the protective capacity of L. salivarius CCFM 1266 against ICB-induced colitis. Importantly, L. salivarius CCFM 1266 did not interfere with the anti-tumor immune response elicited by ipilimumab. Probiotic intervention thus emerges as a promising approach for alleviating ICB-induced colitis.
Collapse
Affiliation(s)
- Shikai Yan
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, China.
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Leilei Yu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, China.
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Fengwei Tian
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, China.
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, China.
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Wei Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, China.
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Haiqin Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, China.
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Qixiao Zhai
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, China.
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| |
Collapse
|
77
|
Liu WX, Liu HN, Weng ZP, Geng Q, Zhang Y, Li YF, Shen W, Zhou Y, Zhang T. Maternal vitamin B1 is a determinant for the fate of primordial follicle formation in offspring. Nat Commun 2023; 14:7403. [PMID: 37973927 PMCID: PMC10654754 DOI: 10.1038/s41467-023-43261-8] [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: 11/14/2022] [Accepted: 11/06/2023] [Indexed: 11/19/2023] Open
Abstract
The mediation of maternal-embryonic cross-talk via nutrition and metabolism impacts greatly on offspring health. However, the underlying key interfaces remain elusive. Here, we determined that maternal high-fat diet during pregnancy in mice impaired preservation of the ovarian primordial follicle pool in female offspring, which was concomitant with mitochondrial dysfunction of germ cells. Furthermore, this occurred through a reduction in maternal gut microbiota-related vitamin B1 while the defects were restored via vitamin B1 supplementation. Intriguingly, vitamin B1 promoted acetyl-CoA metabolism in offspring ovaries, contributing to histone acetylation and chromatin accessibility at the promoters of cell cycle-related genes, enhancement of mitochondrial function, and improvement of granulosa cell proliferation. In humans, vitamin B1 is downregulated in the serum of women with gestational diabetes mellitus. In this work, these findings uncover the role of the non-gamete transmission of maternal high-fat diet in influencing offspring oogenic fate. Vitamin B1 could be a promising therapeutic approach for protecting offspring health.
Collapse
Affiliation(s)
- Wen-Xiang Liu
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock (R2BGL), College of Life Sciences, Inner Mongolia University, Hohhot, 010070, China
| | - Hai-Ning Liu
- Department of Reproductive Medicine, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao, 266011, China
| | - Zhan-Ping Weng
- Department of obstetrical, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao, 266011, China
| | - Qi Geng
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock (R2BGL), College of Life Sciences, Inner Mongolia University, Hohhot, 010070, China
| | - Yue Zhang
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock (R2BGL), College of Life Sciences, Inner Mongolia University, Hohhot, 010070, China
| | - Ya-Feng Li
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock (R2BGL), College of Life Sciences, Inner Mongolia University, Hohhot, 010070, China
| | - Wei Shen
- College of Life Sciences, Institute of Reproductive Sciences, Qingdao Agricultural University, Qingdao, 266109, China
| | - Yang Zhou
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock (R2BGL), College of Life Sciences, Inner Mongolia University, Hohhot, 010070, China.
| | - Teng Zhang
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock (R2BGL), College of Life Sciences, Inner Mongolia University, Hohhot, 010070, China.
| |
Collapse
|
78
|
Wang T, Chen B, Luo M, Xie L, Lu M, Lu X, Zhang S, Wei L, Zhou X, Yao B, Wang H, Xu D. Microbiota-indole 3-propionic acid-brain axis mediates abnormal synaptic pruning of hippocampal microglia and susceptibility to ASD in IUGR offspring. MICROBIOME 2023; 11:245. [PMID: 37932832 PMCID: PMC10629055 DOI: 10.1186/s40168-023-01656-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 08/23/2023] [Indexed: 11/08/2023]
Abstract
BACKGROUND Autism spectrum disorder (ASD) has been associated with intrauterine growth restriction (IUGR), but the underlying mechanisms are unclear. RESULTS We found that the IUGR rat model induced by prenatal caffeine exposure (PCE) showed ASD-like symptoms, accompanied by altered gut microbiota and reduced production of indole 3-propionic acid (IPA), a microbiota-specific metabolite and a ligand of aryl hydrocarbon receptor (AHR). IUGR children also had a reduced serum IPA level consistent with the animal model. We demonstrated that the dysregulated IPA/AHR/NF-κB signaling caused by disturbed gut microbiota mediated the hippocampal microglia hyperactivation and neuronal synapse over-pruning in the PCE-induced IUGR rats. Moreover, postnatal IPA supplementation restored the ASD-like symptoms and the underlying hippocampal lesions in the IUGR rats. CONCLUSIONS This study suggests that the microbiota-IPA-brain axis regulates ASD susceptibility in PCE-induced IUGR offspring, and supplementation of microbiota-derived IPA might be a promising interventional strategy for ASD with a fetal origin. Video Abstract.
Collapse
Affiliation(s)
- Tingting Wang
- Department of Obstetrics, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
| | - Beidi Chen
- Department of Rheumatology and Immunology, Peking University Third Hospital, Beijing, 100191, China
| | - Mingcui Luo
- Department of Obstetrics, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
| | - Lulu Xie
- Department of Pediatrics, Renmin Hospital of Wuhan University, Wuhan, 430071, China
| | - Mengxi Lu
- Department of Obstetrics, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
| | - Xiaoqian Lu
- Department of Pharmacology, Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, 430071, China
| | - Shuai Zhang
- Department of Obstetrics, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
| | - Liyi Wei
- Department of Obstetrics, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
| | - Xinli Zhou
- Department of Pharmacology, Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, 430071, China
| | - Baozhen Yao
- Department of Pediatrics, Renmin Hospital of Wuhan University, Wuhan, 430071, China
| | - Hui Wang
- Department of Pharmacology, Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, 430071, China
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China
| | - Dan Xu
- Department of Obstetrics, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China.
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China.
| |
Collapse
|
79
|
Mancilla VJ, Braden-Kuhle PN, Brice KN, Mann AE, Williams MT, Zhang Y, Chumley MJ, Barber RC, White SN, Boehm GW, Allen MS. A Synthetic Formula Amino Acid Diet Leads to Microbiome Dysbiosis, Reduced Colon Length, Inflammation, and Altered Locomotor Activity in C57BL/6J Mice. Microorganisms 2023; 11:2694. [PMID: 38004705 PMCID: PMC10673175 DOI: 10.3390/microorganisms11112694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 10/23/2023] [Accepted: 10/30/2023] [Indexed: 11/26/2023] Open
Abstract
The effects of synthetic, free-amino acid diets, similar to those prescribed as supplements for (phenylketonuria) PKU patients, on gut microbiota and overall health are not well understood. In the current, multidisciplinary study, we examined the effects of a synthetically-derived, low-fiber, amino acid diet on behavior, cognition, gut microbiome composition, and inflammatory markers. A cohort of 20 male C57BL/6J mice were randomly assigned to either a standard or synthetic diet (n = 10) at post-natal day 21 and maintained for 13 weeks. Sequencing of the 16S rRNA gene from fecal samples revealed decreased bacterial diversity, increased abundance of bacteria associated with disease, such as Prevotella, and a downward shift in gut microbiota associated with fermentation pathways in the synthetic diet group. Furthermore, there were decreased levels of short chain fatty acids and shortening of the colon in mice consuming the synthetic diet. Finally, we measured TNF-α, IL-6, and IL-10 in serum, the hippocampus, and colon, and found that the synthetic diet significantly increased IL-6 production in the hippocampus. These results demonstrate the importance of a multidisciplinary approach to future diet and microbiome studies, as diet not only impacts the gut microbiome composition but potentially systemic health as well.
Collapse
Affiliation(s)
- Viviana J. Mancilla
- Department of Microbiology, Immunology, and Genetics, School of Biomedical Sciences, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Paige N. Braden-Kuhle
- Department of Psychology, College of Science and Engineering, Texas Christian University, Fort Worth, TX 76109, USA
| | - Kelly N. Brice
- Department of Psychology, College of Science and Engineering, Texas Christian University, Fort Worth, TX 76109, USA
| | - Allison E. Mann
- Department of Microbiology, Immunology, and Genetics, School of Biomedical Sciences, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
- Department of Biological Sciences, College of Science, Clemson University, Clemson, SC 29634, USA
| | - Megan T. Williams
- Department of Microbiology, Immunology, and Genetics, School of Biomedical Sciences, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Yan Zhang
- Department of Microbiology, Immunology, and Genetics, School of Biomedical Sciences, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Michael J. Chumley
- Department of Biology, College of Science and Engineering, Texas Christian University, Fort Worth, TX 76109, USA;
| | - Robert C. Barber
- Department of Pharmacology and Neuroscience, School of Biomedical Sciences, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Sabrina N. White
- Department of Microbiology, Immunology, and Genetics, School of Biomedical Sciences, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Gary W. Boehm
- Department of Psychology, College of Science and Engineering, Texas Christian University, Fort Worth, TX 76109, USA
| | - Michael S. Allen
- Department of Microbiology, Immunology, and Genetics, School of Biomedical Sciences, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| |
Collapse
|
80
|
Xiao Y, Li K, Bian J, Zhang Y, Li J, Liu H, Ye Y, Han L, Gong L, Wang M. Urolithin A Protects Neuronal Cells against Stress Damage and Apoptosis by Atp2a3 Inhibition. Mol Nutr Food Res 2023; 67:e2300146. [PMID: 37667442 DOI: 10.1002/mnfr.202300146] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 06/30/2023] [Indexed: 09/06/2023]
Abstract
SCOPE This study aims to investigate the effect and mechanism of Urolithin A (UA) on neuronal stress damage on cognitive impairment in type 2 diabetes mellitus (T2DM) mouse model induced by high-fat diet (HFD) and streptozotocin (STZ). METHODS AND RESULTS T2DM mice fed with UA display an attenuated cognitive impairment along with suppressed endoplasmic reticulum (ER) stress and Tau hyperphosphorylation in brain. Similar restraint effect of UA on Tau hyperphosphorylation and ER stress is also observed in high glucose-treated primary hippocampal neurons. Moreover, UA ameliorates oxidative stress, ER stress, aberrant energy metabolism, and apoptosis in 2,3-dimethoxy-1,4-naphthoquinone (DMNQ) induced HT22 cells. Atp2a3 is identified as a potential target gene of UA which is closely related to intracellular calcium homeostasis, ER stress, and apoptosis, so that UA significantly down-regulated Atp2a3 expression in DMNQ-induced cells. Furthermore, the protection effect of UA against ER stress and apoptosis is abolished by Atp2a3 over-expression in HT22 cells. Taken together, these data suggest that UA performs anti-stress effect by suppressing the expression of Atp2a3 in damaged neuronal cells and thus attenuates diabetes-associated cognitive impairment in T2DM mice. CONCLUSION The study implies UA as a potential novel pharmaceutic target for neurodegeneration and stress damage through regulating the expression of Atp2a3.
Collapse
Affiliation(s)
- Yao Xiao
- College of Food Science and Engineering, Northwest A & F University, Yangling, Shaanxi, 712100, P. R. China
- Department of Orthopaedics, Shanghai Jiao Tong University School of Medicine Affiliated Sixth People's Hospital, 600 Yishan Rd, Shanghai, 200233, P. R. China
| | - Kailin Li
- College of Food Science and Engineering, Northwest A & F University, Yangling, Shaanxi, 712100, P. R. China
| | - Ji Bian
- Kolling Institute, Sydney Medical School, Royal North Shore Hospital, University of Sydney, St. Leonards, NSW, 2065, Australia
| | - Yao Zhang
- College of Food Science and Engineering, Northwest A & F University, Yangling, Shaanxi, 712100, P. R. China
| | - Jia Li
- College of Food Science and Engineering, Northwest A & F University, Yangling, Shaanxi, 712100, P. R. China
| | - Hang Liu
- Shanxi Institute for Functional Food, Shanxi Agricultural University, Taiyuan, Shanxi, 030031, P. R. China
| | - Yingzhi Ye
- Zhejiang Conba Pharmaceutical Co., Ltd, No. 1 Conba Avenue, Lanxi, Zhejiang, 321109, P. R. China
| | - Lin Han
- College of Food Science and Engineering, Northwest A & F University, Yangling, Shaanxi, 712100, P. R. China
| | - Lan Gong
- Microbiome Research Centre, St George and Sutherland Clinical School, University of New South Wales, Sydney, NSW 2052, Australia
| | - Min Wang
- College of Food Science and Engineering, Northwest A & F University, Yangling, Shaanxi, 712100, P. R. China
| |
Collapse
|
81
|
Guo X, Li C, Zhang J, Sun M, Xu J, Xu C, Kuang H, Xu L. Chiral nanoparticle-remodeled gut microbiota alleviates neurodegeneration via the gut-brain axis. NATURE AGING 2023; 3:1415-1429. [PMID: 37946041 DOI: 10.1038/s43587-023-00516-9] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 09/28/2023] [Indexed: 11/12/2023]
Abstract
Alzheimer's disease (AD) is characterized by amyloid-β accumulation in the brain and hyperphosphorylated tau aggregation, as well as neuroinflammation. The gut-brain axis has emerged as a therapeutic target in neurodegenerative diseases by modulating metabolic activity, neuroimmune functions and sensory neuronal signaling. Here we investigate interactions between orally ingested chiral Au nanoparticles and the gut microbiota in AD mice. Oral administration of chiral Au nanoparticles restored cognitive abilities and ameliorated amyloid-β and hyperphosphorylated tau pathologies in AD mice via alterations in the gut microbiome composition and an increase in the gut metabolite, indole-3-acetic acid, which was lower in serum and cerebrospinal fluid of patients with AD compared with age-matched controls. Oral administration of indole-3-acetic acid was able to penetrate the blood-brain barrier and alleviated cognitive decline and pathology including neuroinflammation in AD mice. These findings provide a promising therapeutic target for the amelioration of neuroinflammation and treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Xiao Guo
- State Key Laboratory of Food Science and Resources, International Joint Research Laboratory for Biointerface and Biodetection, School of Food Science and Technology, International Joint Research Center for Photo-responsive Molecules and Materials, Jiangnan University, Wuxi, People's Republic of China
| | - Chen Li
- State Key Laboratory of Food Science and Resources, International Joint Research Laboratory for Biointerface and Biodetection, School of Food Science and Technology, International Joint Research Center for Photo-responsive Molecules and Materials, Jiangnan University, Wuxi, People's Republic of China
| | - Jia Zhang
- State Key Laboratory of Food Science and Resources, International Joint Research Laboratory for Biointerface and Biodetection, School of Food Science and Technology, International Joint Research Center for Photo-responsive Molecules and Materials, Jiangnan University, Wuxi, People's Republic of China
| | - Maozhong Sun
- State Key Laboratory of Food Science and Resources, International Joint Research Laboratory for Biointerface and Biodetection, School of Food Science and Technology, International Joint Research Center for Photo-responsive Molecules and Materials, Jiangnan University, Wuxi, People's Republic of China
| | - Jun Xu
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, China National Clinical Research Center for Neurological Diseases, Beijing, People's Republic of China
| | - Chuanlai Xu
- State Key Laboratory of Food Science and Resources, International Joint Research Laboratory for Biointerface and Biodetection, School of Food Science and Technology, International Joint Research Center for Photo-responsive Molecules and Materials, Jiangnan University, Wuxi, People's Republic of China.
| | - Hua Kuang
- State Key Laboratory of Food Science and Resources, International Joint Research Laboratory for Biointerface and Biodetection, School of Food Science and Technology, International Joint Research Center for Photo-responsive Molecules and Materials, Jiangnan University, Wuxi, People's Republic of China.
| | - Liguang Xu
- State Key Laboratory of Food Science and Resources, International Joint Research Laboratory for Biointerface and Biodetection, School of Food Science and Technology, International Joint Research Center for Photo-responsive Molecules and Materials, Jiangnan University, Wuxi, People's Republic of China.
| |
Collapse
|
82
|
Mort EJ, Heritage S, Jones S, Fowden AL, Camm EJ. Sex-Specific Effects of a Maternal Obesogenic Diet High in Fat and Sugar on Offspring Adiposity, Growth, and Behavior. Nutrients 2023; 15:4594. [PMID: 37960247 PMCID: PMC10648016 DOI: 10.3390/nu15214594] [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: 10/10/2023] [Revised: 10/25/2023] [Accepted: 10/26/2023] [Indexed: 11/15/2023] Open
Abstract
With rising rates of human obesity, this study aimed to determine the relationship between maternal diet-induced obesity, offspring morphometrics, and behavior in mice. Pregnant and lactating female mice fed a diet high in fat and sugar (HFHS) commonly consumed by human populations showed decreased food, calorie, and protein intake but increased adiposity at the expense of lean mass. The pre-weaning body weight of the HFHS offspring was reduced for the first postnatal week but not thereafter, with HFHS female offspring having higher body weights by weaning due to continuing higher fractional growth rates. Post-weaning, there were minor differences in offspring food and protein intake. Maternal diet, however, affected fractional growth rate and total body fat content of male but not female HFHS offspring. The maternal diet did not affect the offspring's locomotor activity or social behavior in either sex. Both the male and female HFHS offspring displayed reduced anxiety-related behaviors, with sex differences in particular aspects of the elevated plus maze task. In the novel object recognition task, performance was impaired in the male but not female HFHS offspring. Collectively, the findings demonstrate that maternal obesity alters the growth, adiposity, and behavior of male and female offspring, with sex-specific differences.
Collapse
Affiliation(s)
- Emily J. Mort
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge CB2 3EG, UK
| | - Sophie Heritage
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge CB2 3EG, UK
| | - Susan Jones
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge CB2 3EG, UK
| | - Abigail L. Fowden
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge CB2 3EG, UK
| | - Emily J. Camm
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge CB2 3EG, UK
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright Street, Clayton, VIC 3168, Australia
| |
Collapse
|
83
|
Li MY, Duan JQ, Wang XH, Liu M, Yang QY, Li Y, Cheng K, Liu HQ, Wang F. Inulin Inhibits the Inflammatory Response through Modulating Enteric Glial Cell Function in Type 2 Diabetic Mellitus Mice by Reshaping Intestinal Flora. ACS OMEGA 2023; 8:36729-36743. [PMID: 37841129 PMCID: PMC10568710 DOI: 10.1021/acsomega.3c03055] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 09/13/2023] [Indexed: 10/17/2023]
Abstract
Inulin, a commonly used dietary fiber supplement, is capable of modulating the gut microbiome. Chronic inflammation resulting from metabolic abnormalities and gut flora dysfunction plays a significant role in the development of type 2 diabetes mellitus (T2DM). Our research has demonstrated that inulin administration effectively reduced colonic inflammation in T2DM mice by inducing changes in the gut microbiota and increasing the concentration of butyric acid, which in turn modulated the function of enteric glial cells (EGCs). Experiments conducted on T2DM mice revealed that inulin administration led to an increase in the Bacteroidetes/Firmicutes ratio and the concentration of butyric acid in the colon. The anti-inflammatory effects of altered gastrointestinal flora and its metabolites were further confirmed through fecal microbiota transplantation. Butyric acid was found to inhibit the activation of the κB inhibitor kinase β/nuclear factor κB pathway, regulate the expression levels of interleukin-6 and tumor necrosis factor-α, suppress the abnormal activation of EGCs, and prevent the release of inflammatory factors by EGCs. Similar results were observed in vitro experiments with butyric acid. Our findings demonstrate that inulin, by influencing the intestinal flora, modifies the activity of EGCs to effectively reduce colonic inflammation in T2DM mice.
Collapse
Affiliation(s)
- Meng-Ying Li
- The
Ministry of Education Key Lab of Hazard Assessment and Control in
Special Operational Environment, The Shaanxi Provincial Key Laboratory
of Environmental Health Hazard Assessment and Protection, The Shaanxi
Provincial Key Laboratory of Free Radical Biology and Medicine, Department
of Health Education and Management, School of Preventive Medicine, Air Force Medical University, West Changle Road No. 169, Xi’an, Shaanxi 710032, China
- Department
of Endocrinology, Xijing Hospital, Air Force
Medical University, West
Changle Road No. 127, Xi’an, Shaanxi 710032, China
| | - Jia-Qi Duan
- The
Ministry of Education Key Lab of Hazard Assessment and Control in
Special Operational Environment, The Shaanxi Provincial Key Laboratory
of Environmental Health Hazard Assessment and Protection, The Shaanxi
Provincial Key Laboratory of Free Radical Biology and Medicine, Department
of Health Education and Management, School of Preventive Medicine, Air Force Medical University, West Changle Road No. 169, Xi’an, Shaanxi 710032, China
| | - Xiao-Hui Wang
- The
Ministry of Education Key Lab of Hazard Assessment and Control in
Special Operational Environment, The Shaanxi Provincial Key Laboratory
of Environmental Health Hazard Assessment and Protection, The Shaanxi
Provincial Key Laboratory of Free Radical Biology and Medicine, Department
of Health Education and Management, School of Preventive Medicine, Air Force Medical University, West Changle Road No. 169, Xi’an, Shaanxi 710032, China
| | - Meng Liu
- School
of Environmental and Municipal Engineering, Xi’an University of Architecture and Technology, Middle of Yanta Road No. 13, Xi’an 710055, China
| | - Qiao-Yi Yang
- The
Ministry of Education Key Lab of Hazard Assessment and Control in
Special Operational Environment, The Shaanxi Provincial Key Laboratory
of Environmental Health Hazard Assessment and Protection, The Shaanxi
Provincial Key Laboratory of Free Radical Biology and Medicine, Department
of Health Education and Management, School of Preventive Medicine, Air Force Medical University, West Changle Road No. 169, Xi’an, Shaanxi 710032, China
| | - Yan Li
- Department
of Anatomy, Histology and Embryology and K. K. Leung Brain Research
Centre, Air Force Medical University, West Changle Road No. 169, Xi’an, Shaanxi 710032, China
| | - Kun Cheng
- Department
of Endocrinology, Xijing Hospital, Air Force
Medical University, West
Changle Road No. 127, Xi’an, Shaanxi 710032, China
| | - Han-Qiang Liu
- The
Ministry of Education Key Lab of Hazard Assessment and Control in
Special Operational Environment, The Shaanxi Provincial Key Laboratory
of Environmental Health Hazard Assessment and Protection, The Shaanxi
Provincial Key Laboratory of Free Radical Biology and Medicine, Department
of Health Education and Management, School of Preventive Medicine, Air Force Medical University, West Changle Road No. 169, Xi’an, Shaanxi 710032, China
| | - Feng Wang
- The
Ministry of Education Key Lab of Hazard Assessment and Control in
Special Operational Environment, The Shaanxi Provincial Key Laboratory
of Environmental Health Hazard Assessment and Protection, The Shaanxi
Provincial Key Laboratory of Free Radical Biology and Medicine, Department
of Health Education and Management, School of Preventive Medicine, Air Force Medical University, West Changle Road No. 169, Xi’an, Shaanxi 710032, China
| |
Collapse
|
84
|
Li F, Chen H, Mao N, Liu H. Dietary fiber intake and cognitive impairment in older patients with chronic kidney disease in the United States: A cross-sectional study. PLoS One 2023; 18:e0291690. [PMID: 37792684 PMCID: PMC10550150 DOI: 10.1371/journal.pone.0291690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Accepted: 09/02/2023] [Indexed: 10/06/2023] Open
Abstract
BACKGROUND High-fiber diet has been associated with better cognitive performance. However, the association between dietary fiber intake and cognition in older patients with chronic kidney disease (CKD) remains unknown. Hence, this study aimed to investigate the effect of dietary fiber intake on cognition in older patients with CKD. METHODS This study included participants aged ≥60 years who provided data on social demography, cognitive tests (Consortium to Establish a Registry for Alzheimer's disease Word Learning [CERAD-WL], CERAD Delayed Recall [CERAD-DR], Animal Fluency Test [AFT], and Digit Symbol Substitution Test [DSST]), diet, and other potential cognition-related variables from the National Health and Nutrition Examination Survey 2011-2014. Fully-adjusted multivariate logistic regression subgroup models were performed, and multiple linear regression analyses were employed to examine the association between dietary fiber intake and cognition in patients with CKD. RESULTS A total of 2461 older adults were included, with 32% who suffered from CKD. Participants with CKD scored lower in CERAD-WL, CERAD-DR, AFT, and DSST. Patients with CKD consuming low dietary fiber (≤25 g/day) had a higher risk of CERAD-WL and DSST impairments. High dietary fiber intake eliminated the differences in CERAD-WL and DSST impairments between the CKD and non-CKD participants. However, no associations were observed between CKD and CERAD-DR and AFT impairments regardless of dietary fiber intake. A positive linear relationship between dietary fiber intake and AFT score was observed in older patients with CKD. CONCLUSION High dietary fiber intake may benefit cognitive function in older patients with CKD. High-fiber diet management strategies could potentially mitigate cognitive impairment in this group of patients.
Collapse
Affiliation(s)
- Feiyan Li
- School of Clinical Medicine, Chengdu Medical College, Chengdu, Sichuan, China
- Department of Experiment Teaching Center of Clinical Medicine, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Hongxi Chen
- Department of Nephrology, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Nan Mao
- Department of Nephrology, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Hong Liu
- School of Clinical Medicine, Chengdu Medical College, Chengdu, Sichuan, China
- Department of Experiment Teaching Center of Clinical Medicine, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| |
Collapse
|
85
|
Tao Q, Zhang ZD, Lu XR, Qin Z, Liu XW, Li SH, Bai LX, Ge BW, Li JY, Yang YJ. Multi-omics reveals aspirin eugenol ester alleviates neurological disease. Biomed Pharmacother 2023; 166:115311. [PMID: 37572635 DOI: 10.1016/j.biopha.2023.115311] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 08/06/2023] [Accepted: 08/07/2023] [Indexed: 08/14/2023] Open
Abstract
BACKGROUND Exosomes play an essential role in maintaining normal brain function due to their ability to cross the blood-brain barrier. Aspirin eugenol ester (AEE) is a new medicinal compound synthesized by the esterification of aspirin with eugenol using the prodrug principle. Aspirin has been reported to have neuroprotective effects and may be effective against neurodegenerative diseases. PURPOSE This study wanted to investigate how AEE affected neurological diseases in vivo and in vitro. EXPERIMENTAL APPROACH A multi-omics approach was used to explore the effects of AEE on the nervous system. Gene and protein expression changes of BDNF and NEFM in SY5Y cells after AEE treatment were detected using RT-qPCR and Western Blot. KEY RESULTS The multi-omics results showed that AEE could regulate neuronal synapses, neuronal axons, neuronal migration, and neuropeptide signaling by affecting transport, inflammatory response, and regulating apoptosis. Exosomes secreted by AEE-treated Caco-2 cells could promote the growth of neurofilaments in SY5Y cells and increased the expression of BDNF and NEFM proteins in SY5Y cells. miRNAs in the exosomes of AEE-treated Caco-2 cells may play an important role in the activation of SY5Y neuronal cells. CONCLUSIONS In conclusion, AEE could play positive effects on neurological-related diseases.
Collapse
Affiliation(s)
- Qi Tao
- Key Lab of New Animal Drug Project of Gansu Province, Key Lab of Veterinary Pharmaceutical Development of Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of CAAS, Lanzhou 730050, China
| | - Zhen-Dong Zhang
- Key Lab of New Animal Drug Project of Gansu Province, Key Lab of Veterinary Pharmaceutical Development of Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of CAAS, Lanzhou 730050, China
| | - Xiao-Rong Lu
- Key Lab of New Animal Drug Project of Gansu Province, Key Lab of Veterinary Pharmaceutical Development of Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of CAAS, Lanzhou 730050, China
| | - Zhe Qin
- Key Lab of New Animal Drug Project of Gansu Province, Key Lab of Veterinary Pharmaceutical Development of Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of CAAS, Lanzhou 730050, China
| | - Xi-Wang Liu
- Key Lab of New Animal Drug Project of Gansu Province, Key Lab of Veterinary Pharmaceutical Development of Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of CAAS, Lanzhou 730050, China
| | - Shi-Hong Li
- Key Lab of New Animal Drug Project of Gansu Province, Key Lab of Veterinary Pharmaceutical Development of Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of CAAS, Lanzhou 730050, China
| | - Li-Xia Bai
- Key Lab of New Animal Drug Project of Gansu Province, Key Lab of Veterinary Pharmaceutical Development of Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of CAAS, Lanzhou 730050, China
| | - Bo-Wen Ge
- Key Lab of New Animal Drug Project of Gansu Province, Key Lab of Veterinary Pharmaceutical Development of Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of CAAS, Lanzhou 730050, China
| | - Jian-Yong Li
- Key Lab of New Animal Drug Project of Gansu Province, Key Lab of Veterinary Pharmaceutical Development of Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of CAAS, Lanzhou 730050, China.
| | - Ya-Jun Yang
- Key Lab of New Animal Drug Project of Gansu Province, Key Lab of Veterinary Pharmaceutical Development of Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of CAAS, Lanzhou 730050, China.
| |
Collapse
|
86
|
Wang L, Wang Z, Lan Y, Tuo Y, Ma S, Liu X. Inulin Attenuates Blood-Brain Barrier Permeability and Alleviates Behavioral Disorders by Modulating the TLR4/MyD88/NF-κB Pathway in Mice with Chronic Stress. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:13325-13337. [PMID: 37642581 DOI: 10.1021/acs.jafc.3c03568] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Depression and vulnerability to chronic stress are associated with inflammatory responses and the loss of blood-brain barrier (BBB) integrity. Dietary fiber and its short-chain fatty acid (SCFAs) metabolites have been reported to affect neuropsychiatric disorders. Here, a 9-week treatment course of inulin (0.037 g of inulin/kcal) exhibited in chronic unpredictable mild stress (CUMS) mice led to antidepressant and anxiolytic effects, as well as improved neurogenesis and synaptic plasticity by enhancing CREB/BDNF signaling. Importantly, inulin inhibited CUMS-induced decreased BBB permeability, reduced lipopolysaccharide (LPS) brain penetration, and modulated TLR4/MyD88/NF-κB signaling to alleviate neuroinflammatory responses. Furthermore, inulin protected the gut barrier integrity and led to the increased formation of SCFAs. Enhanced SCFAs formation was strongly positively correlated with behavioral improvements, BBB integrity, and neuroinflammatory responses. We speculate that dietary fiber may be a promising nutritional intervention to reverse the effects of chronic stress by regulating metabolites and protecting the BBB integrity.
Collapse
Affiliation(s)
- Lei Wang
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China
| | - Zhuo Wang
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China
| | - Yongli Lan
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China
- State Key Laboratory of Plateau Ecology and Agriculture, Qinghai University, Xi'ning 810016, China
| | - Yanliang Tuo
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China
| | - Shaobo Ma
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China
| | - Xuebo Liu
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China
| |
Collapse
|
87
|
Li L, Yang J, Liu T, Shi Y. Role of the gut-microbiota-metabolite-brain axis in the pathogenesis of preterm brain injury. Biomed Pharmacother 2023; 165:115243. [PMID: 37517290 DOI: 10.1016/j.biopha.2023.115243] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 07/09/2023] [Accepted: 07/25/2023] [Indexed: 08/01/2023] Open
Abstract
Brain injury, a common complication in preterm infants, includes the destruction of the key structural and functional connections of the brain and causes neurodevelopmental disorders; it has high morbidity and mortality rates. The exact mechanism underlying brain injury in preterm infants is unclear. Intestinal flora plays a vital role in brain development and the maturation of the immune system in infants; however, detailed understanding of the gut microbiota-metabolite-brain axis in preterm infants is lacking. In this review, we summarise the key mechanisms by which the intestinal microbiota contribute to neurodevelopment and brain injury in preterm infants, with special emphasis on the influence of microorganisms and their metabolites on the regulation of neurocognitive development and neurodevelopmental risks related to preterm birth, infection and neonatal necrotising enterocolitis (NEC). This review provides support for the development and application of novel therapeutic strategies, including probiotics, prebiotics, synbiotics, and faecal bacteria transplantation targeting at brain injury in preterm infants.
Collapse
Affiliation(s)
- Ling Li
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| | - Jiahui Yang
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| | - Tianjing Liu
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| | - Yongyan Shi
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| |
Collapse
|
88
|
Tao Y, Chen W, Xu H, Xu J, Yang H, Luo X, Chen M, He J, Bai Y, Qi H. Adipocyte-Derived Exosomal NOX4-Mediated Oxidative Damage Induces Premature Placental Senescence in Obese Pregnancy. Int J Nanomedicine 2023; 18:4705-4726. [PMID: 37608820 PMCID: PMC10441661 DOI: 10.2147/ijn.s419081] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 08/11/2023] [Indexed: 08/24/2023] Open
Abstract
Background A recent study has reported that maternal obesity is linked to placental oxidative damage and premature senescence. NADPH oxidase 4 (NOX4) is massively expressed in adipose tissue, and its induced reactive oxygen species have been found to contribute to cellular senescence. While, whether, in obese pregnancy, adipose tissue-derived NOX4 is the considerable cause of placental senescence remained elusive. Methods This study collected term placentas from obese and normal pregnancies and obese pregnant mouse model was constructed by a high fat diet to explore placental senescence. Furthermore, adipocyte-derived exosomes were isolated from primary adipocyte medium of obese and normal pregnancies to examine their effect on placenta functions in vivo and vitro. Results The placenta from the obese group showed a significant increase in placental oxidative damage and senescence. Exosomes from obese adipocytes contained copies of NOX4, and when cocultured with HTR8/SVneo cells, they induced severe oxidative damage, cellular senescence, and suppressed proliferation and invasion functions when compared with the control group. In vivo, adipocyte-derived NOX4-containing exosomes could induce placental oxidative damage and senescence, ultimately leading to adverse pregnancy outcomes. Conclusion In obesity, adipose tissue can secrete exosomes containing NOX4 which can be delivered to trophoblast resulting in severe DNA oxidative damage and premature placental senescence, ultimately leading to adverse pregnancy outcomes.
Collapse
Affiliation(s)
- Yuelan Tao
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People’s Republic of China
- Chongqing Key Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, Chongqing, 400016, People’s Republic of China
| | - Wei Chen
- Department of Emergency & Intensive Care Units, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People’s Republic of China
| | - Hongbing Xu
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People’s Republic of China
| | - Jiacheng Xu
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People’s Republic of China
- Chongqing Key Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, Chongqing, 400016, People’s Republic of China
| | - Huan Yang
- Department of Obstetrics, Chongqing University Three Gorges Hospital, Chongqing, 404100, People’s Republic of China
| | - Xin Luo
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People’s Republic of China
| | - Miaomiao Chen
- Maternal and Child Health Hospital of Hubei Province, Wuhan City, Hubei Province, 430070, People’s Republic of China
| | - Jie He
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People’s Republic of China
- Chongqing Key Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, Chongqing, 400016, People’s Republic of China
| | - Yuxiang Bai
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People’s Republic of China
| | - Hongbo Qi
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People’s Republic of China
- Chongqing Key Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, Chongqing, 400016, People’s Republic of China
- Women and Children’s Hospital of Chongqing Medical University, Chongqing, 401147, People’s Republic of China
| |
Collapse
|
89
|
Miyake K, Horiuchi S, Shinohara R, Kushima M, Otawa S, Yui H, Akiyama Y, Ooka T, Kojima R, Yokomichi H, Mochizuki K, Yamagata Z. Maternal dietary fiber intake during pregnancy and child development: the Japan Environment and Children's Study. Front Nutr 2023; 10:1203669. [PMID: 37575329 PMCID: PMC10415901 DOI: 10.3389/fnut.2023.1203669] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 06/20/2023] [Indexed: 08/15/2023] Open
Abstract
Background Animal studies have shown that maternal low-fiber diets during pregnancy may impair brain development and function in offspring, but this has not been validated by epidemiological studies. The aim of this study was to investigate the link between maternal dietary fiber intake during pregnancy and neurodevelopmental delay in offspring using a large birth cohort. Methods A total of 76,207 mother-infant pairs were analyzed using data from the Japan Environment and Children's Study, a nationwide prospective cohort study. Maternal dietary fiber intake was estimated using the food frequency questionnaire in mid-pregnancy. Maternal dietary fiber intake was adjusted for energy and classified into quintiles. Developmental delay was assessed in five domains using the Japanese version of the Ages and Stages Questionnaire, Third Edition at the age of 3 years. The logistic regression analysis was performed to estimate the odds ratio (OR) and 95% confidence interval (CI) for the link between dietary fiber intake during pregnancy and developmental delay at the age of 3 years. Results The lowest intake group of total dietary fiber had a higher risk of delayed communication [adjusted OR (aOR), 1.51; 95% CI, 1.32-1.74], fine motor (aOR, 1.45; 95% CI, 1.32-1.61), problem-solving (aOR, 1.46; 95% CI, 1.32-1.61), and personal-social skills (aOR, 1.30; 95% CI, 1.12-1.50) than did the highest intake group. An analysis that excluded the effects of insufficient folic acid intake during pregnancy also showed a similar trend. Conclusion This study showed that maternal dietary fiber deficiency during pregnancy might influence an increased risk of neurodevelopmental delay in offspring.
Collapse
Affiliation(s)
- Kunio Miyake
- Department of Health Sciences, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Chuo, Japan
| | - Sayaka Horiuchi
- Center for Birth Cohort Studies, University of Yamanashi, Chuo, Japan
| | - Ryoji Shinohara
- Center for Birth Cohort Studies, University of Yamanashi, Chuo, Japan
| | - Megumi Kushima
- Center for Birth Cohort Studies, University of Yamanashi, Chuo, Japan
| | - Sanae Otawa
- Center for Birth Cohort Studies, University of Yamanashi, Chuo, Japan
| | - Hideki Yui
- Department of Health Sciences, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Chuo, Japan
| | - Yuka Akiyama
- Department of Health Sciences, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Chuo, Japan
| | - Tadao Ooka
- Department of Health Sciences, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Chuo, Japan
| | - Reiji Kojima
- Department of Health Sciences, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Chuo, Japan
| | - Hiroshi Yokomichi
- Department of Health Sciences, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Chuo, Japan
| | - Kazuki Mochizuki
- Department of Local Produce and Food Sciences, Faculty of Life and Environmental Sciences, University of Yamanashi, Kofu, Japan
| | - Zentaro Yamagata
- Department of Health Sciences, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Chuo, Japan
- Center for Birth Cohort Studies, University of Yamanashi, Chuo, Japan
| |
Collapse
|
90
|
Deng X, Liang J, Wang L, Niu L, Xiao J, Guo Q, Liu X, Xiao C. Whole Grain Proso Millet ( Panicum miliaceum L.) Attenuates Hyperglycemia in Type 2 Diabetic Mice: Involvement of miRNA Profile. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023. [PMID: 37294881 DOI: 10.1021/acs.jafc.2c08184] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
This work aimed to investigate the hypoglycemic effects and underlying mechanism of whole grain proso millet (Panicum miliaceum L.; WPM) on type 2 diabetes mellitus (T2DM). The results showed that WPM supplementation significantly reduced fasting blood glucose (FBG) and serum lipid levels in T2DM mice induced by a high-fat diet (HFD) combined with streptozotocin (STZ), with improved glucose tolerance, liver and kidney injury, and insulin resistance. In addition, WPM significantly inhibited the expression of gluconeogenesis-related genes G6pase, Pepck, Foxo1, and Pgc-1α. Further study by miRNA high-throughput sequencing revealed that WPM supplementation mainly altered the liver miRNA expression profile of T2DM mice by increasing the expression of miR-144-3p_R-1 and miR-423-5p, reducing the expression of miR-22-5p_R-1 and miR-30a-3p. GO and KEGG analyses showed that the target genes of these miRNAs were mainly enriched in the PI3K/AKT signaling pathway. WPM supplementation significantly increased the level of PI3K, p-AKT, and GSK3β in the liver of T2DM mice. Taken together, WPM exerts antidiabetic effects by improving the miRNA profile and activating the PI3K/AKT signaling pathway to inhibit gluconeogenesis. This study implies that PM can act as a dietary supplement to attenuate T2DM.
Collapse
Affiliation(s)
- Xu Deng
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China
| | - Jiayi Liang
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China
| | - Lehui Wang
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China
| | - Li Niu
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China
| | - Jin Xiao
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China
| | - Qianqian Guo
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China
| | - Xuebo Liu
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China
| | - Chunxia Xiao
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China
| |
Collapse
|
91
|
Yu L, Gao Y, Ye Z, Duan H, Zhao J, Zhang H, Narbad A, Tian F, Zhai Q, Chen W. Interaction of beta-glucans with gut microbiota: Dietary origins, structures, degradation, metabolism, and beneficial function. Crit Rev Food Sci Nutr 2023; 64:9884-9909. [PMID: 37272431 DOI: 10.1080/10408398.2023.2217727] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Beta-glucan (BG), a polysaccharide comprised of interfacing glucose monomers joined via beta-glycosidic linkages, can be defined as a type of dietary fiber with high specificity based on its interaction with the gut microbiota. It can induce similar interindividual microbiota responses, thereby having beneficial effects on the human body. In this paper, we review the four main sources of BG (cereals, fungi, algae, and bacteria) and their differences in structure and content. The interaction of BG with gut microbiota and the resulting health effects have been highlighted, including immune enhancement, regulation of serum cholesterol and insulin levels, alleviation of obesity and improvement of cognitive disorders. Finally, the application of BG in food products and its beneficial effects on the gut microbiota of consumers were discussed. Although some of the mechanisms of action remain unclear, revealing the beneficial functions of BG from the perspective of gut microbiota can help provide theoretical support for the development of diets that target the regulation of microbiota.
Collapse
Affiliation(s)
- Leilei Yu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
- International Joint Research Laboratory for Probiotics, Jiangnan University, Wuxi, Jiangsu, China
| | - Yuhang Gao
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Zi Ye
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Hui Duan
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
- International Joint Research Laboratory for Probiotics, Jiangnan University, Wuxi, Jiangsu, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu, China
| | - Hao Zhang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
- International Joint Research Laboratory for Probiotics, Jiangnan University, Wuxi, Jiangsu, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu, China
| | - Arjan Narbad
- International Joint Research Laboratory for Probiotics, Jiangnan University, Wuxi, Jiangsu, China
- Gut Health and Microbiome Institute Strategic Programme, Quadram Institute Bioscience, Norwich, UK
| | - Fengwei Tian
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
- International Joint Research Laboratory for Probiotics, Jiangnan University, Wuxi, Jiangsu, China
| | - Qixiao Zhai
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
- International Joint Research Laboratory for Probiotics, Jiangnan University, Wuxi, Jiangsu, China
| | - Wei Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
- International Joint Research Laboratory for Probiotics, Jiangnan University, Wuxi, Jiangsu, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu, China
| |
Collapse
|
92
|
Dong B, Qi Y, Sundas H, Yang R, Zhou J, Li Z. Soy protein increases cognitive level in mice by modifying hippocampal nerve growth, oxidative stress, and intestinal microbiota. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2023; 103:4085-4094. [PMID: 36514948 DOI: 10.1002/jsfa.12388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 12/07/2022] [Accepted: 12/14/2022] [Indexed: 05/03/2023]
Abstract
BACKGROUND Three kinds of diet containing chicken protein isolate (CPI), bovine milk protein isolate (BMPI), and soy protein isolate (SPI), respectively, were designed to investigate the influences of proteins on cognitive levels and related mechanisms in mice. RESULTS A Morris water maze (MWM) test showed that the SPI group had a higher cognitive level than the BMPI group. Immunohistochemical staining and chemical analysis of the hippocampus showed that the SPI group had higher synaptophysin expression, doublecortin-positive cell proportion, superoxide dismutase activity, and lower malondialdehyde content compared with the BMPI group. The same parameters in the CPI group were between those of the BMPI and SPI groups. Microbiome sequencing indicated that the three groups differed significantly at the phylum, genus, and species levels, with higher microbial alpha diversity in the CPI and SPI groups. The association of intestinal microbiota with cognitive improvement was also assessed. The present study suggests that soy protein may increase cognitive function by the gut-brain axis. CONCLUSION In contrast with CPI and BMPI, SPI had a better effect on improving the cognitive level in mice, which was achieved through the regulation of hippocampal neural growth, oxidative stress, and intestinal microbiota. © 2022 Society of Chemical Industry.
Collapse
Affiliation(s)
- Beijia Dong
- College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Yuanjin Qi
- College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Hina Sundas
- College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Ruiqi Yang
- College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Jie Zhou
- College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Zhicheng Li
- College of Food Science and Engineering, Northwest A&F University, Yangling, China
| |
Collapse
|
93
|
Cai Y, Liu P, Zhou X, Yuan J, Chen Q. Probiotics therapy show significant improvement in obesity and neurobehavioral disorders symptoms. Front Cell Infect Microbiol 2023; 13:1178399. [PMID: 37249983 PMCID: PMC10213414 DOI: 10.3389/fcimb.2023.1178399] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Accepted: 04/24/2023] [Indexed: 05/31/2023] Open
Abstract
Obesity is a complex metabolic disease, with cognitive impairment being an essential complication. Gut microbiota differs markedly between individuals with and without obesity. The microbial-gut-brain axis is an important pathway through which metabolic factors, such as obesity, affect the brain. Probiotics have been shown to alleviate symptoms associated with obesity and neurobehavioral disorders. In this review, we evaluated previously published studies on the effectiveness of probiotic interventions in reducing cognitive impairment, depression, and anxiety associated with obesity or a high-fat diet. Most of the probiotics studied have beneficial health effects on obesity-induced cognitive impairment and anxiety. They positively affect immune regulation, the hypothalamic-pituitary-adrenal axis, hippocampal function, intestinal mucosa protection, and glucolipid metabolism regulation. Probiotics can influence changes in the composition of the gut microbiota and the ratio between various flora. However, probiotics should be used with caution, particularly in healthy individuals. Future research should further explore the mechanisms underlying the gut-brain axis, obesity, and cognitive function while overcoming the significant variation in study design and high risk of bias in the current evidence.
Collapse
|
94
|
Yuan F, Zhou Z, Wu S, Jiao F, Chen L, Fang L, Yin H, Hu X, Jiang X, Liu K, Xiao F, Jiang H, Chen S, Liu Z, Shu Y, Guo F. Intestinal activating transcription factor 4 regulates stress-related behavioral alterations via paraventricular thalamus in male mice. Proc Natl Acad Sci U S A 2023; 120:e2215590120. [PMID: 37126693 PMCID: PMC10175747 DOI: 10.1073/pnas.2215590120] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 03/31/2023] [Indexed: 05/03/2023] Open
Abstract
Chronic stress induces depression- and anxiety-related behaviors, which are common mental disorders accompanied not only by dysfunction of the brain but also of the intestine. Activating transcription factor 4 (ATF4) is a stress-induced gene, and we previously show that it is important for gut functions; however, the contribution of the intestinal ATF4 to stress-related behaviors is not known. Here, we show that chronic stress inhibits the expression of ATF4 in gut epithelial cells. ATF4 overexpression in the colon relieves stress-related behavioral alterations in male mice, as measured by open-field test, elevated plus-maze test, and tail suspension test, whereas intestine-specific ATF4 knockout induces stress-related behavioral alterations in male mice. Furthermore, glutamatergic neurons are inhibited in the paraventricular thalamus (PVT) of two strains of intestinal ATF4-deficient mice, and selective activation of these neurons alleviates stress-related behavioral alterations in intestinal ATF4-deficient mice. The highly expressed gut-secreted peptide trefoil factor 3 (TFF3) is chosen from RNA-Seq data from ATF4 deletion mice and demonstrated decreased in gut epithelial cells, which is directly regulated by ATF4. Injection of TFF3 reverses stress-related behaviors in ATF4 knockout mice, and the beneficial effects of TFF3 are blocked by inhibiting PVT glutamatergic neurons using DREADDs. In summary, this study demonstrates the function of ATF4 in the gut-brain regulation of stress-related behavioral alterations, via TFF3 modulating PVT neural activity. This research provides evidence of gut signals regulating stress-related behavioral alterations and identifies possible drug targets for the treatment of stress-related behavioral disorders.
Collapse
Affiliation(s)
- Feixiang Yuan
- Zhongshan Hospital, State Key Laboratory of Medical Neurobiology, Institute for Translational Brain Research, Minister of Education Frontiers Center for Brain Science, Fudan University, Shanghai200032, China
| | - Ziheng Zhou
- Chinese Academy of Sciences Key Laboratory of Nutrition, Metabolism and Food Safety, Innovation Center for Intervention of Chronic Disease and Promotion of Health, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai200031, China
| | - Shangming Wu
- Chinese Academy of Sciences Key Laboratory of Nutrition, Metabolism and Food Safety, Innovation Center for Intervention of Chronic Disease and Promotion of Health, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai200031, China
| | - Fuxin Jiao
- Chinese Academy of Sciences Key Laboratory of Nutrition, Metabolism and Food Safety, Innovation Center for Intervention of Chronic Disease and Promotion of Health, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai200031, China
| | - Liang Chen
- Center for Inflammatory Bowel Disease Research, The Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai200072, China
| | - Leilei Fang
- Center for Inflammatory Bowel Disease Research, The Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai200072, China
| | - Hanrui Yin
- Chinese Academy of Sciences Key Laboratory of Nutrition, Metabolism and Food Safety, Innovation Center for Intervention of Chronic Disease and Promotion of Health, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai200031, China
| | - Xiaoming Hu
- Zhongshan Hospital, State Key Laboratory of Medical Neurobiology, Institute for Translational Brain Research, Minister of Education Frontiers Center for Brain Science, Fudan University, Shanghai200032, China
| | - Xiaoxue Jiang
- Zhongshan Hospital, State Key Laboratory of Medical Neurobiology, Institute for Translational Brain Research, Minister of Education Frontiers Center for Brain Science, Fudan University, Shanghai200032, China
| | - Kan Liu
- Chinese Academy of Sciences Key Laboratory of Nutrition, Metabolism and Food Safety, Innovation Center for Intervention of Chronic Disease and Promotion of Health, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai200031, China
| | - Fei Xiao
- Zhongshan Hospital, State Key Laboratory of Medical Neurobiology, Institute for Translational Brain Research, Minister of Education Frontiers Center for Brain Science, Fudan University, Shanghai200032, China
| | - Haizhou Jiang
- Chinese Academy of Sciences Key Laboratory of Nutrition, Metabolism and Food Safety, Innovation Center for Intervention of Chronic Disease and Promotion of Health, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai200031, China
| | - Shanghai Chen
- Zhongshan Hospital, State Key Laboratory of Medical Neurobiology, Institute for Translational Brain Research, Minister of Education Frontiers Center for Brain Science, Fudan University, Shanghai200032, China
| | - Zhanju Liu
- Center for Inflammatory Bowel Disease Research, The Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai200072, China
| | - Yousheng Shu
- Zhongshan Hospital, State Key Laboratory of Medical Neurobiology, Institute for Translational Brain Research, Minister of Education Frontiers Center for Brain Science, Fudan University, Shanghai200032, China
| | - Feifan Guo
- Zhongshan Hospital, State Key Laboratory of Medical Neurobiology, Institute for Translational Brain Research, Minister of Education Frontiers Center for Brain Science, Fudan University, Shanghai200032, China
| |
Collapse
|
95
|
Huang C, Tan H, Song M, Liu K, Liu H, Wang J, Shi Y, Hou F, Zhou Q, Huang R, Shen B, Lin X, Qin X, Zhi F. Maternal Western diet mediates susceptibility of offspring to Crohn's-like colitis by deoxycholate generation. MICROBIOME 2023; 11:96. [PMID: 37131223 PMCID: PMC10155335 DOI: 10.1186/s40168-023-01546-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 04/07/2023] [Indexed: 05/04/2023]
Abstract
BACKGROUND The Western dietary pattern, characterized by high consumption of fats and sugars, has been strongly associated with an increased risk of developing Crohn's disease (CD). However, the potential impact of maternal obesity or prenatal exposure to a Western diet on offspring's susceptibility to CD remains unclear. Herein, we investigated the effects and underlying mechanisms of a maternal high-fat/high-sugar Western-style diet (WD) on offspring's susceptibility to 2,4,6-Trinitrobenzenesulfonic acid (TNBS)-induced Crohn's-like colitis. METHODS Maternal dams were fed either a WD or a normal control diet (ND) for eight weeks prior to mating and continued throughout gestation and lactation. Post-weaning, the offspring were subjected to WD and ND to create four groups: ND-born offspring fed a normal diet (N-N) or Western diet (N-W), and WD-born offspring fed a normal (W-N) or Western diet (W-W). At eight weeks of age, they were administered TNBS to induce a CD model. RESULTS Our findings revealed that the W-N group exhibited more severe intestinal inflammation than the N-N group, as demonstrated by a lower survival rate, increased weight loss, and a shorter colon length. The W-N group displayed a significant increase in Bacteroidetes, which was accompanied by an accumulation of deoxycholic acid (DCA). Further experimentation confirmed an increased generation of DCA in mice colonized with gut microbes from the W-N group. Moreover, DCA administration aggravated TNBS-induced colitis by promoting Gasdermin D (GSDMD)-mediated pyroptosis and IL-1beta (IL-1β) production in macrophages. Importantly, the deletion of GSDMD effectively restrains the effect of DCA on TNBS-induced colitis. CONCLUSIONS Our study demonstrates that a maternal Western-style diet can alter gut microbiota composition and bile acid metabolism in mouse offspring, leading to an increased susceptibility to CD-like colitis. These findings highlight the importance of understanding the long-term consequences of maternal diet on offspring health and may have implications for the prevention and management of Crohn's disease. Video Abstract.
Collapse
Affiliation(s)
- Chongyang Huang
- Guangdong Provincial Key Laboratory of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Gastroenterology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Huishi Tan
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Mengyao Song
- Guangdong Provincial Key Laboratory of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ke Liu
- Guangdong Provincial Key Laboratory of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Hongbin Liu
- Guangdong Provincial Key Laboratory of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jun Wang
- Department of Gastroenterology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yanqiang Shi
- Institute of Dermatology and Venereology, Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Fengyi Hou
- Guangdong Provincial Key Laboratory of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qian Zhou
- Guangdong Provincial Key Laboratory of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ruo Huang
- Guangdong Provincial Key Laboratory of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Binghai Shen
- Guangdong Provincial Key Laboratory of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xinlong Lin
- Guangdong Provincial Key Laboratory of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaoming Qin
- Guangdong Provincial Key Laboratory of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Fachao Zhi
- Guangdong Provincial Key Laboratory of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
96
|
Zhang Q, Bai Y, Wang W, Li J, Zhang L, Tang Y, Yue S. Role of herbal medicine and gut microbiota in the prevention and treatment of obesity. JOURNAL OF ETHNOPHARMACOLOGY 2023; 305:116127. [PMID: 36603782 DOI: 10.1016/j.jep.2022.116127] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 12/16/2022] [Accepted: 12/30/2022] [Indexed: 06/17/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Obesity is a common metabolic dysfunction disease, which is highly correlated with the homeostasis of gut microbiota (GM). The dysregulation of GM on energy metabolism, immune response, insulin resistance and endogenous metabolites (e.g., short chain fatty acids and secondary bile acids) can affect the occurrence and development of obesity. Herbal medicine (HM) has particular advantages and definite therapeutic effects in the prevention and treatment of obesity, but its underlying mechanism is not fully clear. AIM OF THE STUDY In this review, the representative basic and clinical anti-obesity studies associated with the homeostasis of GM regulated by HM including active components, single herb and herbal formulae were summarized and discussed. We aim to provide a state of art reference for the mechanism research of HM in treating obesity and the further development of new anti-obesity drugs. MATERIALS AND METHODS The relevant information was collected by searching keywords (obesity, herbal medicine, prescriptions, mechanism, GM, short chain fatty acids, etc.) from scientific databases (CNKI, PubMed, SpringerLink, Web of Science, SciFinder, etc.). RESULTS GM dysbiosis did occur in obese patients and mice, whiles the intervention of GM could ameliorate the condition of obesity. HM (e.g., berberine, Ephedra sinica, Rehjnannia glutinosa, and Buzhong Yiqi prescription) has been proved to possess a certain regulation on GM and an explicit effect on obesity, but the exact mechanism of HM in improving obesity by regulating GM remains superficial. CONCLUSION GM is involved in HM against obesity, and GM can be a novel therapeutic target for treating obesity.
Collapse
Affiliation(s)
- Qiao Zhang
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi Traditional Chinese Medicine Processing Technology Heritage Base, Shaanxi University of Chinese Medicine, Xi'an, 712046, China.
| | - Yaya Bai
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi Traditional Chinese Medicine Processing Technology Heritage Base, Shaanxi University of Chinese Medicine, Xi'an, 712046, China.
| | - Wenxiao Wang
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi Traditional Chinese Medicine Processing Technology Heritage Base, Shaanxi University of Chinese Medicine, Xi'an, 712046, China.
| | - Jiajia Li
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi Traditional Chinese Medicine Processing Technology Heritage Base, Shaanxi University of Chinese Medicine, Xi'an, 712046, China.
| | - Li Zhang
- Hanlin College, Nanjing University of Chinese Medicine, Taizhou, 225300, Jiangsu Province, China.
| | - Yuping Tang
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi Traditional Chinese Medicine Processing Technology Heritage Base, Shaanxi University of Chinese Medicine, Xi'an, 712046, China.
| | - Shijun Yue
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi Traditional Chinese Medicine Processing Technology Heritage Base, Shaanxi University of Chinese Medicine, Xi'an, 712046, China.
| |
Collapse
|
97
|
Fan P, Wang Y, Lu K, Hong Y, Xu M, Han X, Liu Y. Modeling maternal cholesterol exposure reveals a reduction of neural progenitor proliferation using human cerebral organoids. LIFE MEDICINE 2023; 2:lnac034. [PMID: 39872117 PMCID: PMC11749704 DOI: 10.1093/lifemedi/lnac034] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 08/22/2022] [Indexed: 01/29/2025]
Abstract
Maternal obesity raises the risk of high-cholesterol exposure for their offspring. Studies in cohorts and animal models report that maternal obesity could increase the risk of neurodevelopmental disorders in offspring including intellectual disabilities and autism spectrum disorders (ASDs). However, whether exposure to high cholesterol is responsible for brain developmental defects, as well as its underlying mechanism, is still unclear. Here, we constructed a cholesterol exposure model utilizing human pluripotent stem cell (hPSC)-derived cerebral organoids by exogenously adding cholesterol into the culture system. We observed enlargement of endosomes, decreased neural progenitor proliferation, and premature neural differentiation in brain organoids with the treatment of cholesterol. Moreover, in comparison with published transcriptome data, we found that our single-cell sequencing results showed a high correlation with ASD, indicating that high cholesterol during maternal might mediate the increased risk of ASD in the offspring. Our results reveal a reduction of neural progenitor proliferation in a cholesterol exposure model, which might be a promising indicator for prenatal diagnosis and offer a dynamic human model for maternal environment exposure.
Collapse
Affiliation(s)
- Pan Fan
- Institute for Stem Cell and Neural Regeneration, State Key Laboratory of Reproductive Medicine, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Yuanhao Wang
- Institute for Stem Cell and Neural Regeneration, State Key Laboratory of Reproductive Medicine, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Kaiqin Lu
- Institute for Stem Cell and Neural Regeneration, State Key Laboratory of Reproductive Medicine, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Yuan Hong
- Institute for Stem Cell and Neural Regeneration, State Key Laboratory of Reproductive Medicine, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Min Xu
- Institute for Stem Cell and Neural Regeneration, State Key Laboratory of Reproductive Medicine, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Xiao Han
- Institute for Stem Cell and Neural Regeneration, State Key Laboratory of Reproductive Medicine, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Yan Liu
- Institute for Stem Cell and Neural Regeneration, State Key Laboratory of Reproductive Medicine, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| |
Collapse
|
98
|
Smaga I, Gawlińska K, Gawliński D, Surówka P, Filip M. A maternal high-fat diet during pregnancy and lactation disrupts short-term memory functions via altered hippocampal glutamatergic signaling in female rat offspring. Behav Brain Res 2023; 445:114396. [PMID: 36934986 DOI: 10.1016/j.bbr.2023.114396] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 03/07/2023] [Accepted: 03/16/2023] [Indexed: 03/19/2023]
Abstract
A maternal high-fat diet (HFD) provokes changes in the offspring's brain's structure, function, and development. These changes may cause neuropsychiatric disorders in the early life of offspring the basis of which may be memory impairment. In this study, the effects of maternal HFD during pregnancy and lactation on the short-term memory in adolescent and young adult offspring were evaluated. We analyzed the expression of genes encoding the glutamatergic transporters in the hippocampus to verify the association between changes in glutamatergic transporters and behavioral changes in offspring. Next, we examined whether maternal diet-induced changes in the mRNA levels of genes encoding the NMDA receptor subunits and the AMPA receptor subunits, as well as BDNF in this structure in offspring. All significant changes were validated at the protein level. We found that a maternal HFD during pregnancy and lactation disrupts short-term memory in adolescent and young adult females. The latter change is likely related to the dysregulation of hippocampal levels of GluN2B subunit of NMDA receptors and of reduced levels of BDNF. In summary, we showed that a maternal HFD during pregnancy and lactation triggered several changes within the glutamatergic system in the hippocampus of rat offspring, which may be related to producing behavioral changes in offspring.
Collapse
Affiliation(s)
- Irena Smaga
- Maj Institute of Pharmacology Polish Academy of Sciences, Department of Drug Addiction Pharmacology, Smętna 12, 31-343 Kraków, Poland.
| | - Kinga Gawlińska
- Maj Institute of Pharmacology Polish Academy of Sciences, Department of Drug Addiction Pharmacology, Smętna 12, 31-343 Kraków, Poland
| | - Dawid Gawliński
- Maj Institute of Pharmacology Polish Academy of Sciences, Department of Drug Addiction Pharmacology, Smętna 12, 31-343 Kraków, Poland
| | - Paulina Surówka
- Maj Institute of Pharmacology Polish Academy of Sciences, Department of Drug Addiction Pharmacology, Smętna 12, 31-343 Kraków, Poland
| | - Małgorzata Filip
- Maj Institute of Pharmacology Polish Academy of Sciences, Department of Drug Addiction Pharmacology, Smętna 12, 31-343 Kraków, Poland
| |
Collapse
|
99
|
Lan Y, Ma Z, Chang L, Peng J, Zhang M, Sun Q, Qiao R, Hou X, Ding X, Zhang Q, Peng Q, Dong J, Liu X. Sea buckthorn polysaccharide ameliorates high-fat diet induced mice neuroinflammation and synaptic dysfunction via regulating gut dysbiosis. Int J Biol Macromol 2023; 236:123797. [PMID: 36828095 DOI: 10.1016/j.ijbiomac.2023.123797] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 02/04/2023] [Accepted: 02/18/2023] [Indexed: 02/24/2023]
Abstract
Currently, definitive treatment for neurodegenerative diseases without side effects has not been developed, therefore, exploring natural polysaccharides with neuroprotection to prevent the occurrences and progressions of cognitive dysfunctions has important significance. The purpose of this study was to investigate the effects of sea buckthorn polysaccharide (SBP) on high-fat diet (HFD) induced mice cognitive dysfunctions and attempted to explore its biological mechanisms. Behavior tests (Y-maze and Barnes maze) suggested that SBP effectively alleviated the HFD induced behavioral disorders, which was in accordance with the inhibition of neuroinflammation via suppressing the NF-κB pathway and amelioration of synaptic dysfunction via upregulating CREB/BDNF/TrkB pathway in mice brain. Furthermore, SBP alleviated the gut barrier impairment, inflammatory responses, and lipopolysaccharide invasion into blood circulation via regulating the gut microbiome structure, especially correcting the reduction of Ileibacterium and increase of Lactobacillus, Dubosiella, Olsenella, Helicobacter, and Ruminiclostridium_9 in HFD mice. Therefore, the reversal effects of SBP on gut dysbiosis might be the important reason for its positive effects on cognitive dysfunction induced by HFD in mice.
Collapse
Affiliation(s)
- Ying Lan
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Zhiyuan Ma
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, Shaanxi, China; College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Lili Chang
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Jing Peng
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Mengqi Zhang
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Qingyang Sun
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Ruixue Qiao
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Xinglin Hou
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Xuechao Ding
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Qiang Zhang
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Qiang Peng
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, Shaanxi, China; Puredia Limited, Xining, China
| | - Juane Dong
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, Shaanxi, China; College of Life Sciences, Northwest A&F University, Yangling 712100, Shaanxi, China.
| | - Xuebo Liu
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, Shaanxi, China.
| |
Collapse
|
100
|
Natale F, Spinelli M, Rinaudo M, Cocco S, Nifo Sarrapochiello I, Fusco S, Grassi C. Maternal High Fat Diet Anticipates the AD-like Phenotype in 3xTg-AD Mice by Epigenetic Dysregulation of Aβ Metabolism. Cells 2023; 12:cells12020220. [PMID: 36672155 PMCID: PMC9856666 DOI: 10.3390/cells12020220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/22/2022] [Accepted: 01/03/2023] [Indexed: 01/06/2023] Open
Abstract
Maternal overnutrition has been reported to affect brain plasticity of the offspring by altering gene expression, regulating both synaptic plasticity and adult neurogenesis. However, whether perinatal metabolic stress may influence the accumulation of misfolded proteins and the development of neurodegeneration remains to be clarified. We investigated the impact of maternal high fat diet (HFD) in an experimental model of Alzheimer's disease (AD). The 3xTg-AD mice born to overfed mothers showed an impairment of synaptic plasticity and cognitive deficits earlier than controls. Maternal HFD also altered the expression of genes regulating amyloid-β-protein (Aβ) metabolism (i.e., Bace1, Ern1, Ide and Nicastrin) and enhanced Aβ deposition in the hippocampus. Finally, we found an epigenetic derangement and an aberrant recruitment of transcription factors NF-kB and STAT3 and chromatin remodeler HDAC2 on the regulatory sequences of the same genes. Collectively, our data indicate that early life metabolic stress worsens the AD phenotype via epigenetic alteration of genes regulating Aβ synthesis and clearance.
Collapse
Affiliation(s)
- Francesca Natale
- Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Matteo Spinelli
- Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Marco Rinaudo
- Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Sara Cocco
- Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | | | - Salvatore Fusco
- Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
- Correspondence:
| | - Claudio Grassi
- Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| |
Collapse
|