1
|
Li J, Zhang YJ, Zhao X, Yu Y, Xu JH, Hu R, Wu YH, Huang WQ, Wang ZX, Li TT. Impact of sodium butyrate on stroke-related intestinal injury in diabetic mice: Interference with Caspase-1/GSDMD pyroptosis pathway and preservation of intestinal barrier. Eur J Pharmacol 2025; 998:177455. [PMID: 40057153 DOI: 10.1016/j.ejphar.2025.177455] [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: 07/15/2024] [Revised: 02/27/2025] [Accepted: 03/04/2025] [Indexed: 03/20/2025]
Abstract
Diabetic stroke-associated acute intestinal injury is characterized by high mortality, disability, and poor prognosis due to the lack of effective therapies. Our prior research demonstrated that administration of 300 mg/kg sodium butyrate (NaB) can improve neurological outcomes post-diabetic stroke. Nonetheless, whether the effect of NaB is related to intestinal regulation, along with its underlying mechanisms, remains uncertain. This study aims to investigate the effects and mechanistic pathways of NaB on diabetic stroke-associated acute intestinal injury. A middle cerebral artery occlusion/reperfusion model was established in mice with streptozotocin-induced diabetes. The results demonstrated that NaB alleviated colonic injury 24 h after reperfusion in diabetic stroke. Pyroptosis-related protein levels in colonic tissues were significantly elevated following diabetic stroke but were markedly reduced with NaB treatment. NaB also improved gut barrier integrity and reduced inflammation, promoting epithelial barrier self-repair. In the NaB combined with lipopolysaccharide group, lipopolysaccharide administration induced a significant inflammatory response in the colonic tissue. Conversely, treatment with NaB and VX-765 (an inhibitor for Caspase-1) led to a notable alleviation in intestinal inflammation. These findings suggest that NaB mitigates colonic injury and enhances barrier function following diabetic stroke, potentially through the Caspase-1/Gasdermin D pyroptosis pathway. This study may provide a novel strategy and direction for intestinal rehabilitation in diabetic stroke patients.
Collapse
Affiliation(s)
- Jing Li
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yuan-Jia Zhang
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xu Zhao
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yu Yu
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jing-Hong Xu
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Rong Hu
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ye-Hui Wu
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Wen-Qi Huang
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
| | - Zhong-Xing Wang
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
| | - Ting-Ting Li
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
2
|
Hicks R, Gozal D, Ahmed S, Khalyfa A. Interplay between gut microbiota and exosome dynamics in sleep apnea. Sleep Med 2025; 131:106493. [PMID: 40203611 DOI: 10.1016/j.sleep.2025.106493] [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: 01/12/2025] [Revised: 03/19/2025] [Accepted: 03/29/2025] [Indexed: 04/11/2025]
Abstract
Sleep-disordered breathing (SDB) is characterized by recurrent reductions or interruptions in airflow during sleep, termed hypopneas and apneas, respectively. SDB impairs sleep quality and is linked to substantive health issues including cardiovascular and metabolic disorders, as well as cognitive decline. Recent evidence suggests a link between gut microbiota (GM) composition and sleep apnea. Indeed, GM, a community of microorganisms residing in the gut, has emerged as a potential player in various diseases, and several studies have identified associations between sleep apnea and GM diversity along with shifts in bacterial populations. Additionally, the concept of "leaky gut," a compromised intestinal barrier with potentially increased inflammation, has emerged as another key player in the potential bidirectional relationship between GM and sleep apnea. One of the potential effectors could be extracellular vesicles (EVs) underlying gut-brain communication pathways that are relevant to sleep regulation and function. Thus, therapeutic implications afforded by targeting the GM or exosomes for sleep apnea management have surfaced as promising areas of research. This review explores current understanding of the relationship between GM, exosomes and sleep apnea, highlighting key research dynamics and potential mechanisms. A comprehensive review of the literature was conducted, focusing on studies investigating GM composition, intestinal barrier function and gut-brain communication in relation to sleep apnea.
Collapse
Affiliation(s)
- Rebecca Hicks
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, 25755, USA
| | - David Gozal
- Department of Pediatrics and Office of the Dean, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, 25755, USA
| | - Sarfraz Ahmed
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, 25755, USA
| | - Abdelnaby Khalyfa
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, 25755, USA.
| |
Collapse
|
3
|
Al-Beltagi M. Human milk oligosaccharide secretion dynamics during breastfeeding and its antimicrobial role: A systematic review. World J Clin Pediatr 2025; 14:104797. [PMID: 40491725 PMCID: PMC11947870 DOI: 10.5409/wjcp.v14.i2.104797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 02/19/2025] [Accepted: 02/27/2025] [Indexed: 03/18/2025] Open
Abstract
BACKGROUND Human milk oligosaccharides (HMOs) are bioactive components of breast milk with diverse health benefits, including shaping the gut microbiota, modulating the immune system, and protecting against infections. HMOs exhibit dynamic secretion patterns during lactation, influenced by maternal genetics and environmental factors. Their direct and indirect antimicrobial properties have garnered significant research interest. However, a comprehensive understanding of the secretion dynamics of HMOs and their correlation with antimicrobial efficacy remains underexplored. AIM To synthesize current evidence on the secretion dynamics of HMOs during lactation and evaluate their antimicrobial roles against bacterial, viral, and protozoal pathogens. METHODS A systematic search of PubMed, Scopus, Web of Science, and Cochrane Library focused on studies investigating natural and synthetic HMOs, their secretion dynamics, and antimicrobial properties. Studies involving human, animal, and in vitro models were included. Data on HMO composition, temporal secretion patterns, and mechanisms of antimicrobial action were extracted. Quality assessment was performed using validated tools appropriate for study design. RESULTS A total of 44 studies were included, encompassing human, animal, and in vitro research. HMOs exhibited dynamic secretion patterns, with 2'-fucosyllactose (2'-FL) and lacto-N-tetraose peaking in early lactation and declining over time, while 3-fucosyllactose (3-FL) increased during later stages. HMOs demonstrated significant antimicrobial properties through pathogen adhesion inhibition, biofilm disruption, and enzymatic activity impairment. Synthetic HMOs, including bioengineered 2'-FL and 3-FL, were structurally and functionally comparable to natural HMOs, effectively inhibiting pathogens such as Pseudomonas aeruginosa, Escherichia coli, and Campylobacter jejuni. Additionally, HMOs exhibited synergistic effects with antibiotics, enhancing their efficacy against resistant pathogens. CONCLUSION HMOs are vital in antimicrobial defense, supporting infant health by targeting various pathogens. Both natural and synthetic HMOs hold significant potential for therapeutic applications, particularly in infant nutrition and as adjuncts to antibiotics. Further research, including clinical trials, is essential to address gaps in knowledge, validate findings, and explore the broader applicability of HMOs in improving maternal and neonatal health.
Collapse
Affiliation(s)
- Mohammed Al-Beltagi
- Department of Paediatrics, Faculty of Medicine, Tanta University, Tanta 31511, Alghrabia, Egypt
- Department of Pediatric, University Medical Center, King Abdulla Medical City, Arabian Gulf University, Manama 26671, Bahrain
| |
Collapse
|
4
|
Zeng Q, Hu Y, Xie L, Zhang X, Huang Y, Ye J, Wang S, Xu J. Gut microbiota diversity and composition in children with autism spectrum disorder: associations with symptom severity. PeerJ 2025; 13:e19528. [PMID: 40492208 PMCID: PMC12147763 DOI: 10.7717/peerj.19528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Accepted: 05/06/2025] [Indexed: 06/11/2025] Open
Abstract
Background Autism spectrum disorder (ASD) is a prevalent neurodevelopmental disorder impairing social and communication skills. Gut microbiota has become key in understanding ASD pathophysiology. However, the relationship between the ASD symptoms and alternation of gut microbiota still remains unknow. We hypothesize that the composition of gut microbiota in children with ASD may be strongly associated with the severity of their symptoms. Methods Here, fecal samples from children (divided in to three groups: neurotypical, severe ASD and mild ASD) at a hospital were collected. The symptoms of ASD were assessed by an experienced pediatric neurologist, and the severity of the symptoms in children with ASD was determined based on the assessment scores. Then the diversity and composition of gut microbiota were detected by high-throughput sequencing. Results In total, 2,021 amplicon sequence variants (ASVs) were obtained from 46 fecal samples, with highest in the neurotypical group. Alpha diversity in bacteria differed between severe and mild ASD. Microbiota health and dysbiosis indices varied with ASD severity. Beta diversity indicated that severe ASD differed from others, and mild ASD was closer to neurotypical in community structure. At the phylum level, Firmicutes was the dominant bacteria but abundances differed in different groups, and Ascomycota increased in severe ASD fungi. At the genus level, groups had distinct dominants, and mild ASD microbiota resembled that of neurotypical children. Function prediction revealed differences in bacteria and fungi, with severe ASD having higher amino acid metabolism, lower cofactor/vitamin metabolism, and more undefined saprotrophs. Conclusion This study revealed gut microbiota differences between ASD children (varying symptoms) and neurotypical ones, showing milder ASD closer in microbiota aspects. It offers insights for exploring ASD pathogenesis and devising interventions.
Collapse
Affiliation(s)
- Qinghuang Zeng
- School of Clinical Medicine, Fujian Medical University, Fuzhou, China
- Affiliated Hospital of Putian University, Putian, China
| | - Yisheng Hu
- School of Basic Medicine Science, Key Laboratory of Translational Tumor Medicine in Fujian Province, Putian University, Putian, China
- School of Pharmacy, Putian University, Putian, China
| | - Leiying Xie
- School of Basic Medicine Science, Key Laboratory of Translational Tumor Medicine in Fujian Province, Putian University, Putian, China
| | - Xinyi Zhang
- School of Basic Medicine Science, Key Laboratory of Translational Tumor Medicine in Fujian Province, Putian University, Putian, China
| | - Yun Huang
- School of Basic Medicine Science, Key Laboratory of Translational Tumor Medicine in Fujian Province, Putian University, Putian, China
| | - Jianbin Ye
- School of Basic Medicine Science, Key Laboratory of Translational Tumor Medicine in Fujian Province, Putian University, Putian, China
- School of Pharmacy, Putian University, Putian, China
| | - Shouan Wang
- School of Basic Medicine Science, Key Laboratory of Translational Tumor Medicine in Fujian Province, Putian University, Putian, China
| | - Jia Xu
- School of Basic Medicine Science, Key Laboratory of Translational Tumor Medicine in Fujian Province, Putian University, Putian, China
- School of Basic Medicine Science, Fujian Medical University, Putian, China
| |
Collapse
|
5
|
Liu H, Sun N, Liu Z, Li J, Zhang X. Knockout of bcas3 gene causes neurodevelopment defects in zebrafish. Biol Res 2025; 58:34. [PMID: 40481608 PMCID: PMC12142951 DOI: 10.1186/s40659-025-00615-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Accepted: 05/14/2025] [Indexed: 06/11/2025] Open
Abstract
Background Neurodevelopmental disorders manifest in early childhood and are characterized by cognitive deficits, intellectual disabilities, motor disorders, and social dysfunction. Mutations in BCAS3 gene are associated with syndromic neurodevelopmental disorders in humans, while the detailed pathological mechanism is still unknown. Methods CRISPR/Cas9 technology was used to generate a bcas3 knockout zebrafish model. To investigate the effects of bcas3 on development, morphological evaluations were conducted. Locomotor behaviors, including performance in the light-dark test, novel tank test, mirror test, shoaling test, and social test, were assessed through video tracing and quantitative analysis of movement parameters. Transcriptome sequencing analysis was used to identify dysregulated pathways associated with development process. Additionally, Acridine Orange staining was employed to evaluate apoptosis. Western blot and real-time RT-PCR were used to analyze the expression levels of genes. Results Bcas3 knockout zebrafish exhibited early larval phenotypes resembling clinical features of patients with BCAS3 mutations, including global delayed development at early embryonic development, microcephaly and reduced body length. Behavior analysis revealed abnormal motor dysfunction, such as social impairment, increased anxiety and heightened aggression. Notably, human BCAS3 rescued the developmental defects and motor disorders in bcas3 knockout larvae. Transcriptomic analysis identified substantial downregulation of genes related to embryonic development and startle response, brain development and neuron migration in bcas3 knockout zebrafish, such as rpl10 , cyfip2 , erbb3b , eya4a , nr2f1b , prkg1b and ackr3b . Additionally, increased apoptosis was observed in bcas3 knockout zebrafish, which was further confirmed by Acridine Orange staining and a decreased Bcl2/Bax ratio in western blot analysis. The increased apoptosis observed in the brain of bcas3 knockout larvae could contribute to the developmental and locomotor deficits. Conclusion The bcas3 knockout zebrafish model recapitulates the clinical features observed in patients with BCAS3 mutations. Our results suggest that increased apoptosis may underlie the developmental deficits and motor disorders in these patients. The bcas3 knockout zebrafish model provides a valuable tool to identify dysregulated molecular targets for therapeutic intervention during the early stages of disease progression. Supplementary Information The online version contains supplementary material available at 10.1186/s40659-025-00615-4.
Collapse
Affiliation(s)
- Huihui Liu
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Nianyi Sun
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Zhenxing Liu
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Jinze Li
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Xianqin Zhang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, 430074, China.
| |
Collapse
|
6
|
Zhang X, Xu H, Yin S, Gozal D, Khalyfa A. Obstructive sleep apnea and memory impairments: Clinical characterization, treatment strategies, and mechanisms. Sleep Med Rev 2025; 81:102092. [PMID: 40286536 DOI: 10.1016/j.smrv.2025.102092] [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: 09/25/2024] [Revised: 03/31/2025] [Accepted: 04/07/2025] [Indexed: 04/29/2025]
Abstract
Obstructive sleep apnea (OSA), is associated with dysfunction in the cardiovascular, metabolic and neurological systems. However, the relationship between OSA and memory impairment, intervention effects, and underlying pathways are not well understood. This review summarizes recent advances in the clinical characterization, treatment strategies, and mechanisms of OSA-induced memory impairments. OSA patients may exhibit significant memory declines, including impairments in working memory from visual and verbal sources. The underlying mechanisms behind OSA-related memory impairment are complex and multifactorial with poorly understood aspects that require further investigation. Neuroinflammation, oxidative stress, neuronal damage, synaptic plasticity, and blood-brain barrier dysfunction, as observed under exposures to intermittent hypoxia and sleep fragmentation are likely contributors to learning and memory dysfunction. Continuous positive airway pressure treatment can provide remarkable relief from memory impairment in OSA patients. Other treatments are emerging but need to be rigorously evaluated for cognitive improvement. Clinically, reliable and objective diagnostic tools are necessary for accurate diagnosis and clinical characterization of cognitive impairments in OSA patients. The complex links between gut-brain axis, epigenetic landscape, genetic susceptibility, and OSA-induced memory impairments suggest new directions for research. Characterization of clinical phenotypic clusters can facilitate advances in precision medicine to predict and treat OSA-related memory deficits.
Collapse
Affiliation(s)
- Xiaoman Zhang
- Department of Otolaryngology Head and Neck Surgery & Shanghai Key Laboratory of Sleep Disordered Breathing & Otolaryngology Institute of Shanghai Jiao Tong University, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Huajun Xu
- Department of Otolaryngology Head and Neck Surgery & Shanghai Key Laboratory of Sleep Disordered Breathing & Otolaryngology Institute of Shanghai Jiao Tong University, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
| | - Shankai Yin
- Department of Otolaryngology Head and Neck Surgery & Shanghai Key Laboratory of Sleep Disordered Breathing & Otolaryngology Institute of Shanghai Jiao Tong University, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - David Gozal
- Department of Pediatrics and Office of the Dean, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, 25755, USA
| | - Abdelnaby Khalyfa
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, 25755, USA.
| |
Collapse
|
7
|
Peng Z, Hou T, Yang K, Zhang J, Mao YH, Hou X. Microecologics and Exercise: Targeting the Microbiota-Gut-Brain Axis for Central Nervous System Disease Intervention. Nutrients 2025; 17:1769. [PMID: 40507038 PMCID: PMC12157277 DOI: 10.3390/nu17111769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2025] [Revised: 05/20/2025] [Accepted: 05/22/2025] [Indexed: 06/16/2025] Open
Abstract
The gut microbiota (GM) may play a crucial role in the development and progression of central nervous system (CNS) diseases. Microecologics and exercise can influence the composition and function of GM, thereby exerting positive effects on the CNS. Combined interventions of exercise and microecologics are expected to more comprehensively and effectively address CNS diseases through the microbiota-gut-brain axis (MGBA), potentially outperforming single interventions. However, there is currently a lack of relevant reviews on this topic. In this review, we examine the associations between changes in the microbiota and CNS diseases, including Alzheimer's disease (AD), Parkinson's disease (PD), multiple sclerosis (MS), and autism spectrum disorder (ASD). We also summarize studies on various types of microecologics (such as probiotics, prebiotics, synbiotics, and postbiotics) and exercise in improving CNS disease symptoms. Although current individual studies on microecologics and exercise have achieved certain results, the mechanisms underlying their synergistic effects remain unclear. This review aims to explore the theoretical basis, potential mechanisms, and clinical application prospects of combined interventions of microecologics and exercise in improving CNS diseases through the MGBA, providing a scientific basis for the development of more comprehensive and effective therapeutic interventions.
Collapse
Affiliation(s)
- Zhixing Peng
- School of Exercise and Health, Guangzhou Sport University, Guangzhou 510500, China; (Z.P.); (T.H.); (K.Y.); (J.Z.)
| | - Tingting Hou
- School of Exercise and Health, Guangzhou Sport University, Guangzhou 510500, China; (Z.P.); (T.H.); (K.Y.); (J.Z.)
| | - Keer Yang
- School of Exercise and Health, Guangzhou Sport University, Guangzhou 510500, China; (Z.P.); (T.H.); (K.Y.); (J.Z.)
| | - Jiangyu Zhang
- School of Exercise and Health, Guangzhou Sport University, Guangzhou 510500, China; (Z.P.); (T.H.); (K.Y.); (J.Z.)
| | - Yu-Heng Mao
- School of Exercise and Health, Guangzhou Sport University, Guangzhou 510500, China; (Z.P.); (T.H.); (K.Y.); (J.Z.)
- Guangdong Key Laboratory of Human Sports Performance Science, Guangzhou Sport University, Guangzhou 510500, China
| | - Xiaohui Hou
- School of Exercise and Health, Guangzhou Sport University, Guangzhou 510500, China; (Z.P.); (T.H.); (K.Y.); (J.Z.)
- Guangdong Key Laboratory of Human Sports Performance Science, Guangzhou Sport University, Guangzhou 510500, China
| |
Collapse
|
8
|
Zou S, Yang X, Zhou L. Gut microbiota in epilepsy: How antibiotics induce dysbiosis and influence seizure susceptibility. Microbiol Res 2025; 298:128225. [PMID: 40398011 DOI: 10.1016/j.micres.2025.128225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 04/27/2025] [Accepted: 05/16/2025] [Indexed: 05/23/2025]
Abstract
Epilepsy, a widespread chronic neurological disorder, has recently come under scrutiny for its potential association with the intricate dynamics of gut microbiota. Numerous investigations into the microbiota-gut-brain axis have revealed a close relationship between gut microbiota and epilepsy, suggesting gut microbiota as a potential treatment strategy. In clinical practice, a longstanding correlation has been observed between some kinds of antibiotics and the potential to induce seizures. Consequently, we have conceived a hypothesis that antibiotics might impact seizure activity by modulating the gut microbiota and influencing the physiological processes within the microbiota-gut-brain axis. In this review, our primary objective is to present the existing evidence and theoretical foundations supporting the hypothesis that dysbiosis within the gut microbiota may play a significant role in the pathophysiology of epilepsy. Furthermore, we aim to summarize the possible mechanisms between microbiota-gut-brain axis and epilepsy, offering insights into the selection of appropriate antibiotics for long-term epilepsy management and enhancing therapeutic efficacy through modulation of the gut microbiota. Further research is necessary to fully elucidate the intricate relationship between gut microbiota ecosystem and epilepsy. Exploring these connections holds promise for advancing our understanding of epilepsy pathogenesis and improving patient treatment and care.
Collapse
Affiliation(s)
- Shangnan Zou
- Sun Yat-Sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China; Clinical Neuroscience Center, the Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China.
| | - Xiaofeng Yang
- Guangzhou Medical University, Guangzhou, Guangdong, China; Department of Basic Medicine, Guangzhou National Laboratory, Guangzhou, Guangdong, China.
| | - Liemin Zhou
- Clinical Neuroscience Center, the Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China; Department of Neurology, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China.
| |
Collapse
|
9
|
Zhang J, Liu D, Liu J, Cai C, Hu F, Cheng G, Xu L, Zeng Y. Effects of self-managed lifestyle behavioral changes on cognitive impairment control in Chinese older adults: a population-based prospective study. Transl Psychiatry 2025; 15:165. [PMID: 40360472 PMCID: PMC12075778 DOI: 10.1038/s41398-025-03365-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 03/14/2025] [Accepted: 03/31/2025] [Indexed: 05/15/2025] Open
Abstract
Few studies have examined the effects of self-managed lifestyle behavioral adjustment on cognitive status. This study aimed to explore the association between self-managed behavioral changes and transitions in cognitive status. The Hubei Memory and Aging Cohort Study was a prospective cohort study conducted from 2018-2023 in rural and urban areas. Home-dwelling adults aged ≥65 years completed neuropsychological, lifestyle, clinical, and cognitive assessments. The Cox regressions and cubic splines were used to assess the risk of incident cognitive impairment, and latent class analysis was used to group participants based on behavioral patterns and assess transitions in cognitive status. Among 2477 participants with a mean of 2.02 (SD, 1.25) years of follow-up were included in the study. Participants with low and intermediate compared with high baseline behavioral risk exhibited a reduced risk of incident cognitive impairment. At follow-up, those who maintained stable healthy behaviors or positively adjusted them had a 54% (HR, 0.46 [95% CI, 0.34-0.62]) and 84% (0.16 [0.07-0.35]) lower risk of developing cognitive impairment, respectively, compared with those who maintained unhealthy behaviors. The standard and reinforced behavioral adjustment patterns exhibited a 37% (0.63 [0.22-1.79]) and 77% (0.23 [0.05-0.97]) reduction in the risk of incident cognitive impairment, respectively, compared with the basic pattern. Optimal cognitive gains were attributed to positive adjustments in social networks, physical exercise, cognitive activity, and sleep health. Older adults who maintained healthy behaviors or positively adjusted their unhealthy behaviors exhibited a reduced risk of incident cognitive impairment. Positive behavior modification brought greater cognitive improvement to all participants and more pronounced effects for those with dementia.
Collapse
Affiliation(s)
- Jingjing Zhang
- Hubei Provincial Clinical Research Center for Alzheimer's Disease, Brain Science and Advanced Technology Institute, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
- Geriatric Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, China
- Research Center for Medical AI, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Dan Liu
- Hubei Provincial Clinical Research Center for Alzheimer's Disease, Brain Science and Advanced Technology Institute, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
| | - Jing Liu
- Hubei Provincial Clinical Research Center for Alzheimer's Disease, Brain Science and Advanced Technology Institute, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
| | - Cheng Cai
- Hubei Provincial Clinical Research Center for Alzheimer's Disease, Brain Science and Advanced Technology Institute, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
| | - Feifei Hu
- Hubei Provincial Clinical Research Center for Alzheimer's Disease, Brain Science and Advanced Technology Institute, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
| | - Guirong Cheng
- Hubei Provincial Clinical Research Center for Alzheimer's Disease, Brain Science and Advanced Technology Institute, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
| | - Lang Xu
- Hubei Provincial Clinical Research Center for Alzheimer's Disease, Brain Science and Advanced Technology Institute, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
| | - Yan Zeng
- Hubei Provincial Clinical Research Center for Alzheimer's Disease, Brain Science and Advanced Technology Institute, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China.
- Geriatric Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, China.
| |
Collapse
|
10
|
Wang FX, Dai SY, Mu G, Yu ZH, Chen Y, Zhou J. Beyond organ isolation: The bidirectional crosstalk between cerebral and intestinal ischemia-reperfusion injury via microbiota-gut-brain axis. Biochem Biophys Res Commun 2025; 763:151804. [PMID: 40239544 DOI: 10.1016/j.bbrc.2025.151804] [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: 02/07/2025] [Revised: 03/30/2025] [Accepted: 04/10/2025] [Indexed: 04/18/2025]
Abstract
Ischemia-reperfusion injury (IRI) represents a pathophysiological phenomenon of profound clinical relevance that poses considerable threats to patient safety. IRI may manifest in a variety of clinical contexts including, but not limited to, sepsis, organ transplantation, shock, myocardial infarction, cerebral ischemia, and stroke. Critically, IRI exhibits complex interactions across different organs, with effects that surpass mere localized tissue damage. These impacts can amplify damage to both adjacent and remote organs through pathways such as the gut-brain axis and the gut-lung axis, facilitated by intricate signaling mechanisms. Noteworthy is the interaction between gut IRI and brain IRI, which involves sophisticated neuroendocrine, systemic, and immune mechanisms coordinated through the microbiome-gut-brain axis. This review seeks to delve into the intricate interactions between gut and brain IRI, viewed through the lens of the microbiota-gut-brain axis. It aims to assess its translational potential in clinical settings, provide a theoretical foundation for developing relevant therapeutic strategies, and pinpoint novel directions for research.
Collapse
Affiliation(s)
- Fei-Xiang Wang
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, 646000, China; Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Shi-Yu Dai
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, 646000, China; Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Guo Mu
- Department of Anesthesiology, Zigong Fourth People's Hospital, Zigong, Sichuan, 643000, China
| | - Zi-Hang Yu
- Department of Anesthesiology, Fushun County People's Hospital, Zigong, Sichuan, 643200, China
| | - Ye Chen
- Department of Traditional Chinese Medicine, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Jun Zhou
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, 646000, China; Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Southwest Medical University, Luzhou, Sichuan, 646000, China.
| |
Collapse
|
11
|
Jia S, Mi H, Su Y, Liu Y, Ming Z, Lin J. Changes of intestinal microbiome and its relationship with painful diabetic neuropathy in rats. BMC Microbiol 2025; 25:281. [PMID: 40335921 PMCID: PMC12060437 DOI: 10.1186/s12866-025-04015-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Accepted: 04/30/2025] [Indexed: 05/09/2025] Open
Abstract
OBJECTIVE To analyze the gut bacterial microbiome in rats with painful diabetic neuropathy (PDN) compared to normal rats. METHODS Type 2 diabetes was induced in rats via a high-fat and high-sugar diet combined with a low dose of streptozotocin. Glucose metabolism and insulin sensitivity were evaluated using intraperitoneal glucose tolerance tests and insulin tolerance tests. The progression of peripheral neuropathy was assessed using the mechanical withdrawal threshold and thermal withdrawal latency. Histopathological analysis of rat colon tissues was performed using hematoxylin-eosin staining to observe morphological changes. The expression levels of pro-inflammatory cytokines TNF-α and IL-1β in spinal cord tissues were measured using enzyme-linked immunosorbent assay (ELISA). Fecal samples were then collected for metagenomic sequencing and analysis. RESULT Behavioral tests revealed reduced mechanical withdrawal threshold and thermal withdrawal latency in PDN rats. Histological analysis showed significant colonic mucosal damage and inflammatory cell infiltration, suggesting impaired intestinal barrier function. Elevated TNF-α and IL-1β levels in spinal cord tissues further highlight peripheral inflammation's role in PDN. Sequencing analysis revealed significant differences in gut microbiota composition between PDN and control rats, with altered Bacillota/Bacteroidota ratios and increased Lactobacillus abundance. Functional annotation analysis, based on the KEGG, EggNOG, and CAZy databases, indicated significant enrichment of metabolic pathways related to carbohydrate and amino acid metabolism, energy metabolism, and cell structure biogenesis in PDN rats. Cluster analysis identified higher functional clustering in Metabolism and Genetic Information Processing pathways in PDN rats. CONCLUSION This study demonstrates that PDN leads to altered gut microbiota composition, disrupted metabolic pathways, and increased inflammation, contributing to the pathological progression of diabetic neuropathy. This study provides new insights into the interplay between gut microbiota and diabetic neuropathy, offering potential avenues for therapeutic interventions targeting microbiome and metabolism.
Collapse
Affiliation(s)
- Shuaiying Jia
- Department of Anesthesiology, The Affiliated Hospital of North Sichuan Medical College, 234 Fujiang Road, Shunqing District, Nanchong, Sichuan, 637000, China
| | - Haiqi Mi
- Department of Anesthesiology, The Affiliated Hospital of North Sichuan Medical College, 234 Fujiang Road, Shunqing District, Nanchong, Sichuan, 637000, China
| | - Yao Su
- Department of Anesthesiology, The Affiliated Hospital of North Sichuan Medical College, 234 Fujiang Road, Shunqing District, Nanchong, Sichuan, 637000, China
| | - Yuning Liu
- Department of Anesthesiology, The Affiliated Hospital of North Sichuan Medical College, 234 Fujiang Road, Shunqing District, Nanchong, Sichuan, 637000, China
| | - Zhi Ming
- Department of Anesthesiology, The Affiliated Hospital of North Sichuan Medical College, 234 Fujiang Road, Shunqing District, Nanchong, Sichuan, 637000, China
| | - Jingyan Lin
- Department of Anesthesiology, The Affiliated Hospital of North Sichuan Medical College, 234 Fujiang Road, Shunqing District, Nanchong, Sichuan, 637000, China.
| |
Collapse
|
12
|
Singh S, Saini V, Jha HC. The role of secondary genomes in neurodevelopment and co-evolutionary dynamics. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2025; 180:245-297. [PMID: 40414634 DOI: 10.1016/bs.irn.2025.03.008] [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: 05/27/2025]
Abstract
This chapter examines how human biology and microbial "secondary genomes" have co-evolved to shape neurodevelopment through the gut-brain axis. Microbial communities generate metabolites that cross blood-brain and placental barriers, influencing synaptogenesis, immune responses, and neural circuit formation. Simultaneously, Human Accelerated Regions (HARs) and Endogenous Retroviruses (ERVs) modulate gene expression and immune pathways, determining which microbes thrive in the gut and impacting brain maturation. These factors converge to form a dynamic host-microbe dialogue with significant consequences for neurodevelopmental disorders (NDD), including autism spectrum disorder (ASD), attention-deficit/hyperactivity disorder (ADHD), and schizophrenia. Building on evolutionary perspectives, the chapter elucidates how genetic and immune mechanisms orchestrate beneficial and pathological host-microbe interactions in early brain development. It then explores therapeutic strategies, such as probiotics, prebiotics, fecal microbiota transplantation, and CRISPR-driven microbial engineering, targeting gut dysbiosis to mitigate or prevent neurodevelopmental dysfunctions. Furthermore, innovative organ-on-chip models reveal mechanistic insights under physiologically relevant conditions, offering a translational bridge between in vitro experiments and clinical applications. As the field continues to evolve, this work underscores the translational potential of manipulating the microbiome to optimize neurological outcomes. It enriches our understanding of the intricate evolutionary interplay between host genomes and the microbial world.
Collapse
Affiliation(s)
- Siddharth Singh
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Indore, India.
| | - Vaishali Saini
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Indore, India
| | - Hem Chandra Jha
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Indore, India.
| |
Collapse
|
13
|
Ma M, Li B, Qu Z, Liu S, Li S. Efficacy of probiotics in patients with cognitive impairment: A systematic review and meta-analysis. PLoS One 2025; 20:e0321567. [PMID: 40315198 PMCID: PMC12047807 DOI: 10.1371/journal.pone.0321567] [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: 11/06/2024] [Accepted: 03/09/2025] [Indexed: 05/04/2025] Open
Abstract
OBJECTIVE To conduct an in-depth exploration of the specific impacts of probiotics and prebiotic supplements on cognitive impairment, it is imperative to also investigate pertinent factors, including the optimal dosage of probiotics for enhancing cognitive function. This investigation is essential for optimizing probiotic interventions to prevent and treat cognitive decline, aimed at preventing and aiding in the treatment of cognitive decline among patients with cognitive impairment. METHODS A comprehensive computerized search was conducted across the Embase, PubMed, Web of Science, Cochrane Library, SinoMed, CNKI, Wanfang and WeiPu Data. Studies targeting randomized controlled trials (RCTs) were included. This search covered a timeframe extending from the inception of each database to September 2024. Following an independent process of literature screening, data extraction, and rigorous quality assessment conducted by two investigators, a meta-analysis was performed using Stata 15.0 software. RESULTS A total of ten studies, involving 778 patients, were included in the analysis. The meta-analysis revealed that probiotics were effective in enhancing cognitive function among patients with cognitive impairment, with a standardized mean difference (SMD) of 0.52 (95% CI: 0.07, 0.98; P < 0.001). Subgroup analysis further demonstrated that the largest effect size was observed for studies utilizing the Mini-Mental State Examination (MMSE) scale as the outcome measure (SMD = 0.88). Additionally, the greatest efficacy was associated with single-strain probiotics (SMD = 0.81), and interventions lasting ≤12 weeks exhibited the most pronounced effect (SMD = 0.61). CONCLUSION Probiotics have been shown to enhance cognitive function, with a probiotic intervention program featuring a single probiotic strain and a duration of ≤12 weeks demonstrating particularly robust efficacy in improving cognitive function, as assessed by the MMSE scale.
Collapse
Affiliation(s)
- Miaomiao Ma
- College of Nursing and Health, Henan University, Kaifeng, Henan, China
- Department of Chronic Disease Risk Assessment, Henan University, Kaifeng, Henan, China
| | - Bo Li
- College of Nursing and Health, Henan University, Kaifeng, Henan, China
- Department of Chronic Disease Risk Assessment, Henan University, Kaifeng, Henan, China
| | - Zhi Qu
- College of Nursing and Health, Henan University, Kaifeng, Henan, China
- Department of Chronic Disease Risk Assessment, Henan University, Kaifeng, Henan, China
| | - Shejuan Liu
- College of Nursing and Health, Henan University, Kaifeng, Henan, China
- Department of Chronic Disease Risk Assessment, Henan University, Kaifeng, Henan, China
| | - Sisi Li
- College of Nursing and Health, Henan University, Kaifeng, Henan, China
- Department of Chronic Disease Risk Assessment, Henan University, Kaifeng, Henan, China
| |
Collapse
|
14
|
Inan S, Wilson RP, Tükel Ç. IUPHAR review: From gut to brain: The role of gut dysbiosis, bacterial amyloids, and metabolic disease in Alzheimer's disease. Pharmacol Res 2025; 215:107693. [PMID: 40086611 DOI: 10.1016/j.phrs.2025.107693] [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: 01/17/2025] [Revised: 03/06/2025] [Accepted: 03/07/2025] [Indexed: 03/16/2025]
Abstract
Gut microbial dysbiosis, or altered gut microbial communities, in Alzheimer's Disease suggests a pathogenic role for gut inflammation and microbial products in shaping a neuroinflammatory environment. Similarly, metabolic diseases, such as obesity and diabetes, are also associated with an increased risk of Alzheimer's Disease. As the metabolic landscape shifts during gut inflammation, and gut inflammation in turn impacts metabolic processes, we explore how these interconnected pathways may contribute to the progression of Alzheimer's Disease. Additionally, we discuss the role of bacterial amyloids produced by gut microbes, which may exacerbate amyloid aggregation in the brain and contribute to neurodegenerative processes. Furthermore, we highlight potential therapeutic strategies aimed at reducing gut inflammation, improving metabolic health, and decreasing amyloid content as a means to mitigate Alzheimer's Disease progression. These approaches, targeting the gut-brain-metabolic axis, could offer promising avenues for delaying or preventing cognitive decline in affected individuals.
Collapse
Affiliation(s)
- Saadet Inan
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA.
| | - R Paul Wilson
- Center for Microbiology and Immunology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Çagla Tükel
- Center for Microbiology and Immunology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA.
| |
Collapse
|
15
|
Zhang W, Yi C, Song Z, Yu B, Jiang X, Guo L, Huang S, Xia T, Huang F, Yan Y, Li H, Dai Y. Reshaping the gut microbiota: Tangliping decoction and its core blood-absorbed component quercetin improve diabetic cognitive impairment. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 140:156560. [PMID: 40058319 DOI: 10.1016/j.phymed.2025.156560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 02/11/2025] [Accepted: 02/22/2025] [Indexed: 03/25/2025]
Abstract
BACKGROUND Type 2 diabetes mellitus (T2DM) is associated with an increased risk of cognitive decline, which can result in diabetic cognitive impairment (DCI). Recent studies have indicated that gut microbiota plays a significant role in the development of DCI. Tangliping Decoction (TLP), a traditional Chinese medicine compound, contains various active ingredients that have been shown to regulate the microecology of gut microbiota and potentially improve DCI. However, it remains unclear whether TLP can improve DCI by modulating gut microbiota, as well as which specific component is primarily responsible for these effects. PURPOSE Assess the impact of TLP on alleviating DCI and investigate the contribution of quercetin (QR), the core blood-absorbed component of TLP, in this process. and investigate the underlying mechanisms through which TLP and QR enhance DCI by modulating gut microbiota composition. STUDY DESIGN AND METHODS Initially, experiments such as morris water maze (MWM), morphological analysis, and 16S ribosomal RNA (16S rRNA) gene amplicon sequencing from DCI mice, were performed to validate the pharmacological efficacy of TLP in mitigating DCI. The results indicated that TLP possesses the capacity to modulate the composition and quantity of gut microbiota and safeguard the integrity of the gut barrier and brain barrier. Secondly, high performance liquid chromatography coupled with high-performance liquid chromatography-quadrupole time-of-flight mass spectrometry (HPLC-Q-TOF-MS/MS) combined with network pharmacology methods were used to screen for blood-absorbed components, suggesting that QR may be a potential core blood-absorbed component of TLP in the treatment of DCI. Subsequently, the pharmacological efficacy of QR in ameliorating DCI was confirmed, and the characteristics of gut microbiota as well as the permeability of the gut and brain barrier, were assessed. Finally, fecal microbiota transplantation (FMT) experiments were conducted, wherein fecal matter from TLP and QR-treated mice (donor mice) was transplanted into pseudo-sterile DCI mice with antibiotic-induced depletion of gut microbiota. This approach aimed to elucidate the specific mechanisms by which TLP and QR improve DCI through the modulation of the structure, composition, and abundance of gut microbiota. RESULTS TLP and QR have the potential to enhance learning and memory capabilities in DCI mice, as well as reduce homeostasis model assessment insulin resistance (HOMA-IR) and restore homeostasis model assessment-β function (HOMA- β), leading to increased fasting insulin (FIN) levels and decreased fasting blood glucose (FBG) levels. Simultaneously, the administration of FMT from donor mice to pseudo-sterile DCI mice has been shown to alter the composition and abundance of gut microbiota, leading to amelioration of pathological damage in the colon and hippocampal tissues. Ultimately, FMT utilizing fecal suspensions from donor mice treated with TLP and QR improved cognitive function in pseudo-sterile DCI mice, restore gut microbiota dysbiosis, and maintained the integrity of the gut and brain barriers. CONCLUSION The results of this study indicate that TLP and its core component, QR, which is absorbed into the bloodstream, improve DCI through a gut microbiota-dependent mechanism, providing further evidence for gut microbiota as a therapeutic target for DCI treatment.
Collapse
Affiliation(s)
- Wenlan Zhang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Chunmei Yi
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Zhijie Song
- Department of Rheumatology and Immunology, Chifeng Cancer Hospital, Chifeng 024000, Inner Mongolia Autonomous Region, Chifeng, Inner Mongolia, China
| | - Bin Yu
- School of Acupuncture & Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Xijuan Jiang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Lin Guo
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Shanshan Huang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Tong Xia
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Fayin Huang
- School of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yijing Yan
- School of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Huhu Li
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Yongna Dai
- School of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| |
Collapse
|
16
|
Chen L, Li J, Zhang W, Wang J. Antiepileptic Effects of Acorus tatarinowii Schott in a Rat Model of Epilepsy: Regulation of Metabolic Axes and Gut Microbiota. BIOLOGY 2025; 14:488. [PMID: 40427677 PMCID: PMC12108817 DOI: 10.3390/biology14050488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2025] [Revised: 04/24/2025] [Accepted: 04/28/2025] [Indexed: 05/29/2025]
Abstract
As a phytotherapeutic agent with historical applications in epilepsy management, Acorus tatarinowii Schott (ATS) remains pharmacologically enigmatic, particularly regarding its pathophysiological mechanisms. This knowledge gap significantly hinders the clinical application of ATS-based treatments. To explore the potential of ATS in combating epileptogenesis, we utilized a pentylenetetrazole (PTZ)-induced chronic epilepsy rat model. Brain metabolomic analysis was performed by ultra-performance liquid chromatography coupled with mass spectrometry (UPLC/MS). Principal component analysis (PCA) and orthogonal projections to latent structures-discriminant analysis (OPLS-DA) were performed for screening differential metabolites. Gut microbiota composition was analyzed through 16S rRNA gene sequencing and examined using Spearman correlation analysis. The results show that oral ATS (50 mg/kg) significantly improved the seizure latency and pathology of rats with epilepsy. Ascorbate and aldarate metabolism, glycerophospholipid metabolism, arachidonic acid metabolism, and intestinal flora were crucial for ATS's ability to counteract epilepsy. The therapeutic effects of ATS against epilepsy were investigated with brain metabolomics and gut microbiota analysis, providing the basis for further comprehensive research.
Collapse
Affiliation(s)
- Liang Chen
- School of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China; (L.C.); (J.L.); (W.Z.)
| | - Jiaxin Li
- School of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China; (L.C.); (J.L.); (W.Z.)
| | - Wenhui Zhang
- School of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China; (L.C.); (J.L.); (W.Z.)
| | - Jiepeng Wang
- School of Basic Medical Sciences, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| |
Collapse
|
17
|
Agarwal V, Chaudhary R, Gupta A. Probiotics as a Treatment of Chronic Stress Associated Abnormalities. Probiotics Antimicrob Proteins 2025:10.1007/s12602-025-10553-y. [PMID: 40285929 DOI: 10.1007/s12602-025-10553-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/17/2025] [Indexed: 04/29/2025]
Abstract
Chronic stress is a widespread problem that significantly affects both physical and mental health, leading to numerous complications such as mood disorders, cognitive impairments, gastrointestinal issues, and chronic diseases. The dysregulation of the hypothalamic pituitary adrenal (HPA) axis and the gut-brain axis underlies several stress related disorders, leading to systemic inflammation, neuroinflammation, dysbiosis, and impaired gut barrier integrity. This review emphasizes the growing significance of probiotics as a potential treatment strategy for addressing chronic stress. Probiotics are living bacteria that provide health benefits when consumed in sufficient quantities, acting via several processes including restoration of gut microbial composition, augmentation of gut barrier integrity, and synthesis bioactive compounds such as neurotransmitters and short-chain fatty acids. These effects lead to reduced systemic and neuroinflammation, enhanced neuroplasticity, and the regulation of stress responsive pathways, including the HPA axis. Moreover, probiotics enhance parasympathetic nervous system activity by modulating vagus signaling. Current review indicates the promise of probiotics in alleviating chronic stress; nonetheless, substantial gaps exist regarding strain specific benefits, appropriate doses, and long-term safety. It is essential to address these constraints by comprehensive, large scale clinical studies and tailored therapies. This review highlights the significance of probiotics as a natural, non-invasive approach to chronic stress management, providing an innovative solution for the worldwide issue of stress related health problems.
Collapse
Affiliation(s)
- Vipul Agarwal
- MIT College of Pharmacy, Ram Ganga Vihar Phase-II, Moradabad, 244001, U.P, India.
| | - Rishabh Chaudhary
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University (A Central University), Vidya Vihar, Raebareli Road, Lucknow, 226 025, U.P, India
| | - Anugya Gupta
- Faculty of Medical and Paramedical Sciences, Madhyanchal Professional University, Bhopal, 462044, M.P, India.
| |
Collapse
|
18
|
Zhao H, Qiu X, Wang S, Wang Y, Xie L, Xia X, Li W. Multiple pathways through which the gut microbiota regulates neuronal mitochondria constitute another possible direction for depression. Front Microbiol 2025; 16:1578155. [PMID: 40313405 PMCID: PMC12043685 DOI: 10.3389/fmicb.2025.1578155] [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: 02/17/2025] [Accepted: 03/31/2025] [Indexed: 05/03/2025] Open
Abstract
As a significant mental health disorder worldwide, the treatment of depression has long faced the challenges of a low treatment rate, significant drug side effects and a high relapse rate. Recent studies have revealed that the gut microbiota and neuronal mitochondrial dysfunction play central roles in the pathogenesis of depression: the gut microbiota influences the course of depression through multiple pathways, including immune regulation, HPA axis modulation and neurotransmitter metabolism. Mitochondrial function serves as a key hub that mediates mood disorders through mechanisms such as defective energy metabolism, impaired neuroplasticity and amplified neuroinflammation. Notably, a bidirectional regulatory network exists between the gut microbiota and mitochondria: the flora metabolite butyrate enhances mitochondrial biosynthesis through activation of the AMPK-PGC1α pathway, whereas reactive oxygen species produced by mitochondria counteract the flora composition by altering the intestinal epithelial microenvironment. In this study, we systematically revealed the potential pathways by which the gut microbiota improves neuronal mitochondrial function by regulating neurotransmitter synthesis, mitochondrial autophagy, and oxidative stress homeostasis and proposed the integration of probiotic supplementation, dietary fiber intervention, and fecal microbial transplantation to remodel the flora-mitochondrial axis, which provides a theoretical basis for the development of novel antidepressant therapies targeting gut-brain interactions.
Collapse
Affiliation(s)
- Hongyi Zhao
- School of Basic Medical Science, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiongfeng Qiu
- School of Basic Medical Science, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Shuyu Wang
- School of Health Preservation and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yi Wang
- School of Basic Medical Science, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Li Xie
- School of Basic Medical Science, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiuwen Xia
- School of Basic Medical Science, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Weihong Li
- School of Basic Medical Science, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Sichuan College of Traditional Chinese Medicine, Mianyang, China
| |
Collapse
|
19
|
Xu Z, Chen H, Zheng G, Zhang W, Wang J, Zhang J, Gao Q, Wu J, Xue X. Plasma trimethylamine N-oxide, carnitine, choline, and betaine distinguish cognition impairment in Parkinson's disease. Parkinsonism Relat Disord 2025; 135:107844. [PMID: 40286463 DOI: 10.1016/j.parkreldis.2025.107844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2025] [Revised: 03/30/2025] [Accepted: 04/15/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND Trimethylamine N-oxide (TMAO) and its precursors, as intestinal metabolites, have already been explored in Parkinson's disease (PD). However, whether such an association reflects the risk of cognitive impairment (CI) remains unclear. The purpose of this research is to analyze the diagnostic value of TMAO and its precursors in the detection of PD-related CI. METHODS Using high-performance liquid chromatography-tandem mass spectrometry, plasma TMAO and its precursors were detected from 220 healthy controls (HC) and 234 PD patients who underwent cognitive function assessment. 162 PD patients were enrolled in normal cognitive function group (PD-NCI), while 72 in cognitive impairment group (PD-CI). The expression levels of TMAO and its precursors were compared across different groups. The research carried out logistic regression analysis and built receiver operating characteristic (ROC) curves to assess the diagnostic utility of TMAO and its precursors. RESULTS Compared to HC, there were higher expression levels of TMAO and precursor carnitine and betaine in the PD and PD-CI groups (p < 0.001). A higher plasma TMAO level exists in PD-CI compared to that of PD-NCI (p < 0.001). After adjusting for differentiating variables, logistic regression analysis substantiated the findings. The combination of TMAO and its precursors demonstrated fine predictive value with high sensitivity and specificity with areas under the ROC curve (AUCs) of 0.880, 0.707, and 0.941 for distinguishing between PD and HC, PD-NCI and PD-CI, and PD-CI and HC, respectively. CONCLUSION TMAO and its metabolic precursors are potential biomarkers for early identification of cognitive dysfunction, offering timely opportunities for intervention in PD patients.
Collapse
Affiliation(s)
- Zhaohan Xu
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, Jiangsu, China
| | - Hucheng Chen
- Department of Nuclear Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, Jiangsu, China
| | - Guoyan Zheng
- Department of Nuclear Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, Jiangsu, China
| | - Wei Zhang
- Department of Nuclear Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, Jiangsu, China
| | - Jing Wang
- Department of Nuclear Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, Jiangsu, China
| | - Jian Zhang
- Department of Nuclear Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, Jiangsu, China
| | - Qing Gao
- Department of Nuclear Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, Jiangsu, China; Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, Jiangsu, China.
| | - Jun Wu
- Department of Clinical Laboratory, Nanjing Brain Hospital & the Affiliated Brain Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China.
| | - Xue Xue
- Department of Nuclear Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, Jiangsu, China.
| |
Collapse
|
20
|
Qiu X, Zhang R, Cheng Y, Jin C, Zhang Y, Zhang Z. Oral health implications in Parkinson's disease. NPJ Parkinsons Dis 2025; 11:73. [PMID: 40216783 PMCID: PMC11992149 DOI: 10.1038/s41531-025-00927-y] [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: 10/06/2024] [Accepted: 03/27/2025] [Indexed: 04/14/2025] Open
Abstract
To assist patients in maintaining oral health in patients with Parkinson's disease (PD), this review aims to search the literature on aspects related to oral health in PD patients. In contrast to other research, we included the involvement of microbes in PD patients' poor oral health. Finally, we conclude that, in comparison to healthy individuals, PD patients have poorer oral health and a higher incidence of oral ailments.
Collapse
Affiliation(s)
- Xiaohui Qiu
- School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, 110002, China
| | - Ran Zhang
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, 110002, China
| | - Yi Cheng
- School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, 110002, China
| | - Chengde Jin
- School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, 110002, China
| | - Yushi Zhang
- School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, 110002, China
| | - Ziqi Zhang
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, 110002, China.
| |
Collapse
|
21
|
Li S, Li J, Chen K, Wang J, Wang L, Feng C, Wang K, Xu Y, Gao Y, Yan X, Zhao Q, Li B, Qiu Y. Chronic Arsenic Exposure Causes Alzheimer's Disease Characteristic Effects and the Intervention of Fecal Microbiota Transplantation in Rats. J Appl Toxicol 2025. [PMID: 40204291 DOI: 10.1002/jat.4782] [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: 01/17/2025] [Revised: 03/25/2025] [Accepted: 03/28/2025] [Indexed: 04/11/2025]
Abstract
Arsenic exposure and intestinal microbiota disorders may be related with Alzheimer's disease (AD), but the mechanism has not been elucidated. This study conducted chronic arsenic exposure from rat's maternal body to adult offspring to investigate the mechanisms of the characteristic effects of chronic arsenic exposure on AD, and further explored the intervention effect of fecal microbiota transplantation (FMT) on arsenic-mediated neurotoxicity. Transmission electron microscopy, HE staining, and related indicators were measured in the control group, the exposed group, and the FMT intervention group. Western blot was used to determine microtubule-associated proteins Tau and p-Tau396, intestinal-brain barrier-related proteins Claudin-1 and Occludin, ELISA was used to detect the content of Aβ1-42, and 16S rRNA sequencing was used to detect the intestinal flora of feces. Results showed that chronic arsenic exposure could lead to neurobehavioral defects in rats, increase the expression levels of Tau, p-Tau396, and Aβ1-42 in hippocampus (p < 0.05), increase the abundance of Clostridium _ UCG-014, decrease the abundance of Roseburia, and decrease the expression levels of Claudin-1 and Occludin in colon and hippocampus (p < 0.05). After FMT intervention, the expression levels of Tau and p-Tau396 were decreased (p < 0.05), and the abundance of Roseburia was increased. In summary, chronic arsenic exposure caused intestinal flora disorder by changing the abundance of inflammation-related flora, thereby destroying the gut-brain barrier and causing AD characteristic effects in rats. Although the bacterial specific genus was improved and the expression of AD-related proteins was reduced after transplantation, it could not alleviate the neurobehavioral defects and neurotoxicity caused by arsenic exposure.
Collapse
Affiliation(s)
- Shuyuan Li
- Department of Toxicology, School of Public Health, Shanxi Medical University, Jinzhong, Shanxi, China
| | - Jia Li
- Department of Toxicology, School of Public Health, Shanxi Medical University, Jinzhong, Shanxi, China
| | - Kun Chen
- Department of Toxicology, School of Public Health, Shanxi Medical University, Jinzhong, Shanxi, China
| | - Jing Wang
- Department of Toxicology, School of Public Health, Shanxi Medical University, Jinzhong, Shanxi, China
| | - Longmei Wang
- Department of Toxicology, School of Public Health, Shanxi Medical University, Jinzhong, Shanxi, China
| | - Chao Feng
- Department of Toxicology, School of Public Health, Shanxi Medical University, Jinzhong, Shanxi, China
| | - Kanglin Wang
- Department of Toxicology, School of Public Health, Shanxi Medical University, Jinzhong, Shanxi, China
| | - Yifan Xu
- Department of Toxicology, School of Public Health, Shanxi Medical University, Jinzhong, Shanxi, China
| | - Yi Gao
- Department of Toxicology, School of Public Health, Shanxi Medical University, Jinzhong, Shanxi, China
- Key Laboratory of Coal Environmental Pathogenicity and Prevention, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Key Laboratory of Environmental Health Impairment and Prevention, Shanxi Medical University, Taiyuan, Shanxi, China
- Center for Ecological Public Health Security of Yellow River Basin, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xiaoyan Yan
- Department of Toxicology, School of Public Health, Shanxi Medical University, Jinzhong, Shanxi, China
- Key Laboratory of Coal Environmental Pathogenicity and Prevention, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Key Laboratory of Environmental Health Impairment and Prevention, Shanxi Medical University, Taiyuan, Shanxi, China
- Center for Ecological Public Health Security of Yellow River Basin, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Qian Zhao
- Department of Toxicology, School of Public Health, Shanxi Medical University, Jinzhong, Shanxi, China
- Key Laboratory of Coal Environmental Pathogenicity and Prevention, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Key Laboratory of Environmental Health Impairment and Prevention, Shanxi Medical University, Taiyuan, Shanxi, China
- Center for Ecological Public Health Security of Yellow River Basin, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Ben Li
- Department of Toxicology, School of Public Health, Shanxi Medical University, Jinzhong, Shanxi, China
- Key Laboratory of Coal Environmental Pathogenicity and Prevention, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Key Laboratory of Environmental Health Impairment and Prevention, Shanxi Medical University, Taiyuan, Shanxi, China
- Center for Ecological Public Health Security of Yellow River Basin, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yulan Qiu
- Department of Toxicology, School of Public Health, Shanxi Medical University, Jinzhong, Shanxi, China
- Key Laboratory of Coal Environmental Pathogenicity and Prevention, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Key Laboratory of Environmental Health Impairment and Prevention, Shanxi Medical University, Taiyuan, Shanxi, China
- Center for Ecological Public Health Security of Yellow River Basin, Shanxi Medical University, Taiyuan, Shanxi, China
| |
Collapse
|
22
|
Eslami M, Adampour Z, Fadaee Dowlat B, Yaghmayee S, Motallebi Tabaei F, Oksenych V, Naderian R. A Novel Frontier in Gut-Brain Axis Research: The Transplantation of Fecal Microbiota in Neurodegenerative Disorders. Biomedicines 2025; 13:915. [PMID: 40299512 PMCID: PMC12025253 DOI: 10.3390/biomedicines13040915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2025] [Revised: 04/02/2025] [Accepted: 04/07/2025] [Indexed: 04/30/2025] Open
Abstract
The gut-brain axis (GBA) represents a sophisticated bidirectional communication system connecting the central nervous system (CNS) and the gastrointestinal (GI) tract. This interplay occurs primarily through neuronal, immune, and metabolic pathways. Dysbiosis in gut microbiota has been associated with multiple neurodegenerative diseases, such as Parkinson's disease (PD), Alzheimer's disease (AD), multiple sclerosis (MS), and amyotrophic lateral sclerosis (ALS). In recent years, fecal microbiota transplantation (FMT) has gained attention as an innovative therapeutic approach, aiming to restore microbial balance in the gut while influencing neuroinflammatory and neurodegenerative pathways. This review explores the mechanisms by which FMT impacts the gut-brain axis. Key areas of focus include its ability to reduce neuroinflammation, strengthen gut barrier integrity, regulate neurotransmitter production, and reinstate microbial diversity. Both preclinical and clinical studies indicate that FMT can alleviate motor and cognitive deficits in PD and AD, lower neuroinflammatory markers in MS, and enhance respiratory and neuromuscular functions in ALS. Despite these findings, several challenges remain, including donor selection complexities, uncertainties about long-term safety, and inconsistencies in clinical outcomes. Innovations such as synthetic microbial communities, engineered probiotics, and AI-driven analysis of the microbiome hold the potential to improve the precision and effectiveness of FMT in managing neurodegenerative conditions. Although FMT presents considerable promise as a therapeutic development, its widespread application for neurodegenerative diseases requires thorough validation through well-designed, large-scale clinical trials. It is essential to establish standardized protocols, refine donor selection processes, and deepen our understanding of the molecular mechanisms behind its efficacy.
Collapse
Affiliation(s)
- Majid Eslami
- Cancer Research Center, Semnan University of Medical Sciences, Semnan 35147-99442, Iran;
- Department of Bacteriology and Virology, Faculty of Medicine, Semnan University of Medical Sciences, Semnan 35147-99442, Iran
| | - Zarifeh Adampour
- Institute of Science, Biotechnology and Biosafety Department, Eskishehir Osmangazi University, Eskishehir 26040, Türkiye;
| | - Bahram Fadaee Dowlat
- School of Medicine, Iran University of Medical Sciences, Tehran 14496-14535, Iran
| | - Shayan Yaghmayee
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan 35147-99442, Iran
| | - Faezeh Motallebi Tabaei
- Department of Medical Microbiology, Faculty of Medicine, Golestan University of Medical Sciences, Gorgan 49189-36316, Iran
| | | | - Ramtin Naderian
- Clinical Research Development Unit, Kowsar Educational, Research and Therapeutic Hospital, Semnan University of Medical Sciences, Semnan 35147-99442, Iran
| |
Collapse
|
23
|
Zhu H, Huang C, Luo Z, Wu L, Cheng X, Wu H. Porphyromonas gingivalis Induces Disturbance of Kynurenine Metabolism Through the Gut-Brain Axis: Implications for Alzheimer's Disease. J Dent Res 2025; 104:439-448. [PMID: 39905278 DOI: 10.1177/00220345241303141] [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] [Indexed: 02/06/2025] Open
Abstract
Porphyromonas gingivalis is one of the major pathogens of chronic periodontitis. P. gingivalis can cause systemic inflammation, amyloid β protein deposition, and hyperphosphorylation of tau protein, leading to Alzheimer's disease (AD)-like lesions. P. gingivalis oral infection causes gut microbiota alteration, gut barrier dysfunction, and intestinal immune response and inflammation. The microbiota-gut-brain axis has a potential role in the pathogenesis of AD. Whether P. gingivalis affects AD-like lesions via the gut-brain axis needs more study. In this study, orally administered P. gingivalis induced alveolar resorption, intestinal barrier impairment, and AD-like lesions. Oral infection with P. gingivalis induced oral and gut microflora dysbiosis, imbalance of the tryptophan metabolism pathway of gut microbiota, and elevated levels of 3-hydroxykynurenine in the sera and hippocampi. The key metabolite, 3-hydroxykynurenine, suppressed Bcl2 gene expression, leading to neuronal apoptosis and promoting AD-like lesions in vivo and in vitro. These findings suggest that P. gingivalis can induce AD pathogenesis through the gut-brain axis, providing new ideas for the prevention and treatment of AD.
Collapse
Affiliation(s)
- H Zhu
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Geriatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - C Huang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Geriatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Z Luo
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Geriatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - L Wu
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - X Cheng
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Geriatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - H Wu
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Geriatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
24
|
Zhou W, Zhou Y, Zhang S, Li B, Li Z, Bai Z, Sun D, Huangfu C, Wang N, Xia T, Huang C, Guan L, Yang X, Hu Y, Zhang P, Shen P, Wang R, Ni Z, Gao Y. Gut microbiota's role in high-altitude cognitive impairment: the therapeutic potential of Clostridium sp. supplementation. SCIENCE CHINA. LIFE SCIENCES 2025; 68:1132-1148. [PMID: 39704932 DOI: 10.1007/s11427-024-2779-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 11/13/2024] [Indexed: 12/21/2024]
Abstract
Prolonged exposure to high-altitude environments may increase the risk of cognitive decline in young migrants. Recent studies suggest that hypobaric hypoxia-induced alterations in gut microbial composition could partly contribute to this risk. However, the absence of direct evidence from cohort studies and an unclear mechanism hinder intervention development based on this hypothesis. This study recruited 109 young male migrants living in Xizang to investigate the microbial mechanisms underlying cognitive impairment associated with high-altitude migration. Multi-omic analysis revealed distinct microbiome and metabolome features in migrants with cognitive decline, notably a reduced abundance of Clostridium species and disrupted fecal absorption of L-valine. Mechanistic studies showed that hypobaric hypoxia significantly damaged the intestinal barrier, leading to lipopolysaccharide (LPS) leakage and an influx of inflammatory factors into the peripheral blood, which activated microglia and caused neuronal injury in the hippocampus of mice. Additionally, compromised L-valine absorption due to intestinal barrier damage correlated with lower hippocampal glutamate levels and neurotrophic factors. Intervention with Clostridium sp. effectively restored the intestinal barrier and enhanced L-valine absorption, which mitigated hypobaric hypoxia-induced inflammation and hippocampal neural damage in mice. In conclusion, cognitive impairment among young migrants at high altitude may be attributed to hypobaric hypoxia-induced gut microbiota disruption and subsequent intestinal barrier dysfunction. This study may provide a promising approach for preventing and treating high-altitude-associated cognitive impairment.
Collapse
Affiliation(s)
- Wei Zhou
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Yongqiang Zhou
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Shikun Zhang
- Department of Stem Cell and Regenerative Medicine, Institute of Health Service and Transfusion Medicine, Beijing, 100850, China
| | - Bin Li
- Mountain Sickness Research Institute, No.950 Hospital, Yecheng, 844900, China
| | - Zhong Li
- Department of Stomatology, the First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Zhijie Bai
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Dezhi Sun
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Chaoji Huangfu
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Ningning Wang
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Tiantian Xia
- Medical School of Qinghai University, Xining, 810016, China
| | - Congshu Huang
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Lina Guan
- General Hospital of Xinjiang Military Command, Urumqi, 830000, China
| | - Xi Yang
- General Hospital of Xinjiang Military Command, Urumqi, 830000, China
| | - Yangyi Hu
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Pengfei Zhang
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Pan Shen
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, 100850, China.
| | - Rui Wang
- General Hospital of Xinjiang Military Command, Urumqi, 830000, China.
| | - Zhexin Ni
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, 100850, China.
| | - Yue Gao
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, 100850, China.
- State Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, Beijing, 100853, China.
| |
Collapse
|
25
|
Zhao R, Lu H, Yuan H, Chen S, Xu K, Zhang T, Liu Z, Jiang Y, Suo C, Chen X. Plasma proteomics-based organ-specific aging for all-cause mortality and cause-specific mortality: a prospective cohort study. GeroScience 2025; 47:1411-1423. [PMID: 39477866 PMCID: PMC11978558 DOI: 10.1007/s11357-024-01411-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 10/23/2024] [Indexed: 04/09/2025] Open
Abstract
Individual's aging rates vary across organs. However, there are few methods for assessing aging at organ levels and whether they contribute differently to mortalities remains unknown. We analyzed data from 45,821 adults in the UK Biobank, using plasma proteomics and machine learning to estimate biological ages for 12 major organs. The differences between biological age and chronological age, referred to as "age gaps," were calculated for each organ. Partial correlation analyses were used to assess the association between age gaps and modifiable factors. Adjusted multivariable Cox regression models were applied to examine the association of age gaps with all-cause mortality, cause-specific mortalities, and cancer-specific mortalities. We reveal a complex network of varied associations between multi-organ aging and modifiable factors. All age gaps increase the risk of all-cause mortality by 6-60%. The risk of death varied from 5.54 to 29.18 times depending on the number of aging organs. Cause-specific mortalities are associated with certain organs' aging. For mental diseases mortality, and nervous system mortality, only brain aging exhibited a significant increased risk of HR 2.38 (per SD, 95% CI: 2.06-2.74) and 1.99 (per SD, 95% CI: 1.84-2.16), respectively. Age gaps of stomach were also a specific indicator for gastric cancer. Eventually, we find that an organ's biological age selectively influences the aging of other organ systems. Our study demonstrates that accelerated aging in specific organs increases the risk of mortality from various causes. This provides a potential tool for early identification of at-risk populations, offering a relatively objective method for precision medicine.
Collapse
Affiliation(s)
- Renjia Zhao
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Zhangjiang Fudan International Innovation Center, and School of Life Science, Fudan University, Songhu Road 2005, Shanghai, China
| | - Heyang Lu
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Huangbo Yuan
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Zhangjiang Fudan International Innovation Center, and School of Life Science, Fudan University, Songhu Road 2005, Shanghai, China
| | - Shuaizhou Chen
- Department of Epidemiology and Ministry of Education Key Laboratory of Public Health Safety, School of Public Health, Fudan University, Shanghai, China
| | - Kelin Xu
- Department of Biostatistics, School of Public Health, Fudan University, Shanghai, China
- Fudan University Taizhou Institute of Health Sciences, Taizhou, Jiangsu, China
| | - Tiejun Zhang
- Department of Epidemiology and Ministry of Education Key Laboratory of Public Health Safety, School of Public Health, Fudan University, Shanghai, China
- Department of Biostatistics, School of Public Health, Fudan University, Shanghai, China
- Fudan University Taizhou Institute of Health Sciences, Taizhou, Jiangsu, China
| | - Zhenqiu Liu
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Zhangjiang Fudan International Innovation Center, and School of Life Science, Fudan University, Songhu Road 2005, Shanghai, China
- Fudan University Taizhou Institute of Health Sciences, Taizhou, Jiangsu, China
| | - Yanfeng Jiang
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Zhangjiang Fudan International Innovation Center, and School of Life Science, Fudan University, Songhu Road 2005, Shanghai, China
- Fudan University Taizhou Institute of Health Sciences, Taizhou, Jiangsu, China
| | - Chen Suo
- Department of Epidemiology and Ministry of Education Key Laboratory of Public Health Safety, School of Public Health, Fudan University, Shanghai, China.
- Fudan University Taizhou Institute of Health Sciences, Taizhou, Jiangsu, China.
- Shanghai Institute of Infectious Disease and Biosecurity, Shanghai, China.
| | - Xingdong Chen
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Zhangjiang Fudan International Innovation Center, and School of Life Science, Fudan University, Songhu Road 2005, Shanghai, China.
- Fudan University Taizhou Institute of Health Sciences, Taizhou, Jiangsu, China.
- Yiwu Research Institute of Fudan University, Yiwu, Zhejiang, China.
- National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
26
|
Xiao QX, Qin HY, Chen JJ, Fang CL, Wang QL, Li QJ, Zhu SY, Xiong LL. Multi-omics analysis reveals the potential mechanisms underlying long-term exercise-induced enhancement of learning and memory in male mice. Biogerontology 2025; 26:84. [PMID: 40159584 DOI: 10.1007/s10522-025-10225-y] [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: 10/26/2024] [Accepted: 03/20/2025] [Indexed: 04/02/2025]
Abstract
Exercise is widely recognized for improving physical functions, learning, and memory. However, the mechanisms behind these effects are not fully understood. This study aims to investigate the potential mechanisms through which exercise enhances learning and memory in mice using multi-omics analysis. Twenty male C57BL/6J mice were divided into exercise and control groups. The exercise group underwent a 4-month treadmill training regimen (12 m/min). Learning and memory abilities were assessed using the Morris water maze test. Brain, serum, and fecal samples were collected for neurotransmitter analysis, serum metabolomics analysis, and gut microbiota analysis. Data from neurotransmitter and serum metabolomics analyses were integrated with gut microbiota analysis. For comparisons between the two groups, the independent sample t-test was employed. For comparisons involving multiple groups, a repeated measures one-way analysis of variance (ANOVA) with random unit group design was applied. Statistical significance was defined as P < 0.05. The Morris water maze test significantly improved learning and memory in the exercise group (P < 0.05). Neurotransmitter analysis revealed significant differences in cognitive function-related neurotransmitters and pathways between the exercise and control groups (P < 0.05). Serum metabolomics analysis identified differences in serum metabolites between the two groups, which were linked to key pathways involved in neural repair and cognitive function. Microbial sequencing showed greater gut microbiota diversity in the exercise group, with the most notable changes at the genus level, particularly in Allobaculum, A2, and Clostridium_sensu_stricto_1 (P < 0.05). Integrated analysis indicated correlations between changes in gut microbiota and serum metabolites associated with cognitive function. Long-term exercise enhances learning and memory in mice through multiple mechanisms, including neurotransmitter regulation, serum metabolite changes, and modulation of gut microbiota. These findings provide new insights into the neuroprotective effects of exercise.
Collapse
Affiliation(s)
- Qiu-Xia Xiao
- Department of Anesthesiology, The Third Affiliated Hospital of Zunyi Medical University (The First People's Hospital of Zunyi), Zunyi, 563000, Guizhou, China
| | - Hao-Yue Qin
- Department of Anesthesiology, The Third Affiliated Hospital of Zunyi Medical University (The First People's Hospital of Zunyi), Zunyi, 563000, Guizhou, China
| | - Jun-Jie Chen
- Animal Zoology Department, Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Chang-Le Fang
- Department of Anesthesiology, The Third Affiliated Hospital of Zunyi Medical University (The First People's Hospital of Zunyi), Zunyi, 563000, Guizhou, China
| | - Qiu-Lin Wang
- Department of Anesthesiology, The Third Affiliated Hospital of Zunyi Medical University (The First People's Hospital of Zunyi), Zunyi, 563000, Guizhou, China
| | - Qi-Jun Li
- Department of Anesthesiology, The Third Affiliated Hospital of Zunyi Medical University (The First People's Hospital of Zunyi), Zunyi, 563000, Guizhou, China
| | - Shuai-Yu Zhu
- Department of Anesthesiology, The Third Affiliated Hospital of Zunyi Medical University (The First People's Hospital of Zunyi), Zunyi, 563000, Guizhou, China
| | - Liu-Lin Xiong
- Department of Anesthesiology, The Third Affiliated Hospital of Zunyi Medical University (The First People's Hospital of Zunyi), Zunyi, 563000, Guizhou, China.
| |
Collapse
|
27
|
Yang Z, Zhang Y, Ran S, Zhang J, Tian F, Shi H, Wei S, Li X, Li X, Gao Y, Jia G, Lin H, Chen Z, Zhang Z. A Multi-Omics Study of Neurodamage Induced by Growth-Stage Real-Time Air Pollution Exposure in Mice via the Microbiome-Gut-Brain Axis. TOXICS 2025; 13:260. [PMID: 40278577 PMCID: PMC12031392 DOI: 10.3390/toxics13040260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 03/25/2025] [Accepted: 03/27/2025] [Indexed: 04/26/2025]
Abstract
Air pollution has been widely recognized as a risk factor for neurological disorders, and the gut microbiome may play a mediating role. However, current evidence remains limited. In this study, a mouse model was employed with continuous exposure to real-time air pollution from conception to late adolescence. Effects of growth-stage air pollution exposure on the gut microbiome, host metabolites, and brain tissue were assessed. Pathological damage in the hippocampus and cortex was observed. Fecal metagenomic sequencing revealed alterations in both compositions and functions of the gut microbiome. Metabolic disturbances in unsaturated fatty acids and glycerophospholipids were identified in the intestine, serum, and brain tissues, with significant changes in metabolites (e.g., gamma-linolenic acid, alpha-linolenic acid, docosahexaenoic acid (DHA), phosphatidylethanolamine (PE), phosphatidylcholine (PC) and phosphatidylserine (PS). Serum levels of the pro-inflammatory mediator leukotriene C4 were also elevated. Correlation analysis identified a group of different gut microbiome species that were associated with host metabolites. Furthermore, mediation analysis showed that intestinal and serum metabolites mediated the associations between the key gut microbiome and brain microbiome. These findings indicate that the metabolic crosstalk in the gut-brain axis mediates the neuronal damage in mice induced by growth-stage air pollution exposure, potentially through pathways involving lipid metabolism and inflammation.
Collapse
Affiliation(s)
- Zijun Yang
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China; (Z.Y.); (S.R.); (J.Z.); (F.T.); (H.S.); (S.W.); (X.L.); (Y.G.); (H.L.)
| | - Yi Zhang
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, Beijing 100191, China; (Y.Z.); (G.J.)
| | - Shanshan Ran
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China; (Z.Y.); (S.R.); (J.Z.); (F.T.); (H.S.); (S.W.); (X.L.); (Y.G.); (H.L.)
| | - Jingyi Zhang
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China; (Z.Y.); (S.R.); (J.Z.); (F.T.); (H.S.); (S.W.); (X.L.); (Y.G.); (H.L.)
| | - Fei Tian
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China; (Z.Y.); (S.R.); (J.Z.); (F.T.); (H.S.); (S.W.); (X.L.); (Y.G.); (H.L.)
| | - Hui Shi
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China; (Z.Y.); (S.R.); (J.Z.); (F.T.); (H.S.); (S.W.); (X.L.); (Y.G.); (H.L.)
| | - Shengtao Wei
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China; (Z.Y.); (S.R.); (J.Z.); (F.T.); (H.S.); (S.W.); (X.L.); (Y.G.); (H.L.)
| | - Xiuxiu Li
- Department of Science and Education, Nanshan Maternity & Child Healthcare Hospital of Shenzhen, Shenzhen 518067, China;
| | - Xinyue Li
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China; (Z.Y.); (S.R.); (J.Z.); (F.T.); (H.S.); (S.W.); (X.L.); (Y.G.); (H.L.)
| | - Yonggui Gao
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China; (Z.Y.); (S.R.); (J.Z.); (F.T.); (H.S.); (S.W.); (X.L.); (Y.G.); (H.L.)
| | - Guang Jia
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, Beijing 100191, China; (Y.Z.); (G.J.)
| | - Hualiang Lin
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China; (Z.Y.); (S.R.); (J.Z.); (F.T.); (H.S.); (S.W.); (X.L.); (Y.G.); (H.L.)
| | - Zhangjian Chen
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, Beijing 100191, China; (Y.Z.); (G.J.)
| | - Zilong Zhang
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China; (Z.Y.); (S.R.); (J.Z.); (F.T.); (H.S.); (S.W.); (X.L.); (Y.G.); (H.L.)
| |
Collapse
|
28
|
Dehghani E, Karimi K, Arekhi S, Ardeshir M, Rezapour R, Shayestehfar M, Memari AH. Effect of nutritional supplements on gut microbiome in individuals with neurodevelopmental disorders: a systematic review and narrative synthesis. BMC Nutr 2025; 11:64. [PMID: 40158118 PMCID: PMC11954342 DOI: 10.1186/s40795-025-01043-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 03/12/2025] [Indexed: 04/01/2025] Open
Abstract
BACKGROUND Neurodevelopmental disorders (NDDs) encompass a range of disruptive conditions with varying prevalence rates and multiple contributing factors. Recent studies have suggested a potential connection between NDDs and the gut-brain axis. Furthermore, there is evidence indicating that nutritional supplements might have an impact on gastrointestinal (GI) and behavioral symptoms. This study aimed to explore the effects of nutritional supplements on the gut microbiota and behavioral symptoms in individuals with NDDs. METHODS A systematic search of databases such as PubMed, Scopus, Web of Science, Embase, and APA PsycINFO was conducted, utilizing relevant keywords until February 2025. In addition, the search for gray literature was carried out on Google Scholar and ProQuest. The risk of bias was assessed using the ROBINS-I tool for non-randomized studies and the RoB-1 tool for randomized controlled trials. Due to the heterogeneity of the studies, a Synthesis without Meta-analysis (SWiM) approach was employed. RESULTS The overall findings from the studies indicated positive effects of supplementation in reducing the Gastrointestinal Severity Index (GIS) score and alleviating GI symptoms. Supplementation with probiotics and vitamins increased good microbiomes (GM) and decrease in bad microbiomes (BM) among individuals with autism spectrum disorder (ASD). Moreover, the Firmicutes to Bacteroidetes ratio (F/R ratio) exhibited significant changes after supplementation. Additionally, improvements were observed in various assessment scores, including ATEC, ABC, CARS, and PGI-2. CONCLUSIONS Nutritional supplementation in individuals with NDDs can have a positive influence by modulating the microbiome, reducing dysbiosis, and enhancing gut barrier integrity. Shifting in the F/R ratio can be considered as the reason for improving gastrointestinal and behavioral symptoms by influencing neurotransmitter activity and neuroinflammation. Targeting the gut-brain axis with interventions that focus on gut microbiota offers a promising adjunct therapy for the management of NDD. Registration of the review protocol. PROSPERO registration no. CRD42023460449.
Collapse
Affiliation(s)
- Elaheh Dehghani
- Department of Clinical Nutrition, School of Nutrition and Dietetics, Tehran University of Medical Sciences (TUMS), Tehran, Iran
- Sports Medicine Research Center (SMRC), Neuroscience Institute, Tehran University of Medical Sciences (TUMS), Tehran, Iran
- Nutrition and Metabolic Diseases Research Center, Clinical Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences (AJUMS), Ahvaz, Iran
| | - Keyvan Karimi
- Sports Medicine Research Center (SMRC), Neuroscience Institute, Tehran University of Medical Sciences (TUMS), Tehran, Iran
- Department of Epidemiology and Biostatistics, School of Public Health, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Soheil Arekhi
- Sports Medicine Research Center (SMRC), Neuroscience Institute, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Maryam Ardeshir
- Iranian National Center for Addiction Studies, Tehran University of Medical Sciences, Tehran (TUMS), Tehran, Iran
| | - Reshad Rezapour
- Sports Medicine Research Center (SMRC), Neuroscience Institute, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Monir Shayestehfar
- Sports Medicine Research Center (SMRC), Neuroscience Institute, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Amir Hossein Memari
- Sports Medicine Research Center (SMRC), Neuroscience Institute, Tehran University of Medical Sciences (TUMS), Tehran, Iran.
| |
Collapse
|
29
|
Qi F, Meng K, Zhao X, Lv J, Huang L, Fan X, Feng Z. Targeting gut microbiota: a potential therapeutic approach for tumor microenvironment in glioma. Front Neurol 2025; 16:1549465. [PMID: 40183013 PMCID: PMC11965986 DOI: 10.3389/fneur.2025.1549465] [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: 12/21/2024] [Accepted: 03/03/2025] [Indexed: 04/05/2025] Open
Abstract
Glioma, being one of the malignant tumors with the highest mortality rate globally, has an unclear pathogenesis, and the existing treatment effects still have certain limitations. The tumor microenvironment (TME) plays an important role in the occurrence, development, and recurrence of glioma. As one of the important regulatory factors of TME, the gut microbiota can regulate the progression of glioma not only by interacting with the brain through the brain-gut axis but also by influencing the tumor immune microenvironment (TIME) and inflammatory microenvironment. Recent studies have identified the gut microbiota and TME as potential therapeutic targets for glioma. This paper aims to summarize the role of the gut microbiota in TME, the association between them and glioma, and the potential of developing new intervention measures by targeting the gut microbiota. Understanding the involvement process of the gut microbiota in glioma may pave the way for the development of effective treatment methods that can regulate TME and prevent disease progression.
Collapse
Affiliation(s)
- Fan Qi
- College of Integrated Traditional and Western Medicine, Shaanxi University of Chinese Medicine, Shaanxi, China
| | - Kaiqiang Meng
- College of Traditional Chinese Medicine, Shaanxi University of Chinese Medicine, Shaanxi, China
| | - Xiaoping Zhao
- Neurosurgery Department of the Encephalopathy Hospital, Affiliated Hospital of Shaanxi University of Chinese Medicine, Shaanxi, China
| | - Jing Lv
- College of Integrated Traditional and Western Medicine, Shaanxi University of Chinese Medicine, Shaanxi, China
| | - Lan Huang
- College of Integrated Traditional and Western Medicine, Shaanxi University of Chinese Medicine, Shaanxi, China
| | - Xiaoxuan Fan
- College of Integrated Traditional and Western Medicine, Shaanxi University of Chinese Medicine, Shaanxi, China
| | - Zhaoqun Feng
- Neurosurgery Department of the Encephalopathy Hospital, Affiliated Hospital of Shaanxi University of Chinese Medicine, Shaanxi, China
| |
Collapse
|
30
|
Dong H, Zang C, Liu L, Guo L, Ye X, Li X, Zhou C, Sun C, Yang M, Wei X, Lin B, Li H, Wang H, Qi Y, Hu H, Li N. Lung-specific SFTPC mutations lead to neurodevelopmental disorders with neuroinflammation. Biochem Biophys Res Commun 2025; 753:151479. [PMID: 39965266 DOI: 10.1016/j.bbrc.2025.151479] [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: 02/04/2025] [Revised: 02/06/2025] [Accepted: 02/10/2025] [Indexed: 02/20/2025]
Abstract
Neurodevelopmental disorders (NDDs) are characterized by diverse genetic underpinnings and abnormalities in the structure and function of the central nervous system. While the lung-specific SFTPC gene is critical for pulmonary development and homeostasis, its potential involvement in NDDs has not been previously explored. In this study, we identified compound heterozygous variants of SFTPC in two children diagnosed with NDDs, inherited from carrier parents. Bioinformatic analyses predicted these variants to be deleterious, and patient blood samples confirmed reduced SFTPC protein levels. To investigate the functional impact of these mutations, we generated a Sftpc-knock-in (Sftpc-KI) mouse model carrying the defective alleles. The Sftpc-KI mice exhibited significantly reduced Sftpc expression in both lung and blood samples. Remarkably, despite its lung-specific expression, Sftpc-KI mice displayed pronounced impairments in neurobehavioral performance. Proteomic analyses of the Sftpc-KI mouse brain revealed dysregulated proteins associated with neuroinflammation. Furthermore, primary microglial cells isolated from these mice exhibited heightened expression of M1 activation markers, indicating aberrant microglial activation. Our findings uncover a previously unrecognized connection between lung-specific SFTPC dysfunction and neurodevelopmental disorders, suggesting the existence of a novel brain-lung axis and opening new avenues for research into the molecular mechanisms underlying NDDs.
Collapse
Affiliation(s)
- Haipeng Dong
- Department of Child Health Care, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 510623, Guangzhou, China
| | - Congwen Zang
- Laboratory of Medical Systems Biology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 510623, Guangzhou, China
| | - Lili Liu
- Laboratory of Medical Systems Biology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 510623, Guangzhou, China
| | - Leqin Guo
- Department of Child Health Care, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 510623, Guangzhou, China
| | - Xiangyan Ye
- Laboratory of Medical Systems Biology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 510623, Guangzhou, China
| | - Xiangmiao Li
- Laboratory of Medical Systems Biology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 510623, Guangzhou, China
| | - Chang Zhou
- Laboratory of Medical Systems Biology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 510623, Guangzhou, China
| | - Chuanbo Sun
- Laboratory of Medical Systems Biology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 510623, Guangzhou, China
| | - Miaomiao Yang
- Laboratory of Medical Systems Biology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 510623, Guangzhou, China
| | - Xinshu Wei
- Laboratory of Medical Systems Biology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 510623, Guangzhou, China
| | - Bing Lin
- Laboratory of Medical Systems Biology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 510623, Guangzhou, China
| | - Hong Li
- Laboratory of Medical Systems Biology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 510623, Guangzhou, China
| | - Hanhong Wang
- Laboratory of Medical Systems Biology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 510623, Guangzhou, China
| | - Yifei Qi
- Laboratory of Medical Systems Biology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 510623, Guangzhou, China
| | - Hao Hu
- Laboratory of Medical Systems Biology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 510623, Guangzhou, China; Third Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, China.
| | - Na Li
- Laboratory of Medical Systems Biology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 510623, Guangzhou, China.
| |
Collapse
|
31
|
Chen G, Li Y, Wei S, Wang X, Kuang Z, Guo W, Qin J, Huang T, Li Y, Zhu C. Role of gut microbiota in thalassemia: a review of therapeutic prospects. Front Physiol 2025; 16:1523448. [PMID: 40177354 PMCID: PMC11962020 DOI: 10.3389/fphys.2025.1523448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 03/05/2025] [Indexed: 04/05/2025] Open
Abstract
In recent years, the study of gut microbiota has gradually become a research hotspot in the field of medicine, as gut microbiota dysbiosis is closely related to various diseases. Thalassemia, as a hereditary hemoglobinopathy, has a complex pathophysiological mechanism, and traditional treatment methods show limited efficacy. With a deeper understanding of the gut microbiome, researchers have begun to focus on its role in the pathogenesis of thalassemia and its therapeutic effects. This article aims to review the role of gut microbiota in thalassemia and its potential therapeutic prospects, analyze the latest research findings, and explore the impact and mechanisms of gut microbiota on patients with thalassemia, with the goal of providing new ideas and directions for future research and clinical treatment of thalassemia.
Collapse
Affiliation(s)
- Guanjun Chen
- Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, China
| | - Yulan Li
- Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, China
| | - Shirui Wei
- Shandong Second Medical University, Weifang, Shandong, China
| | - Xinyu Wang
- Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, China
| | - Zheshu Kuang
- Chenzhou Third People’s Hospital (Group), Chenzhou, Hunan, China
| | - Weiming Guo
- Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, China
| | - Jianbin Qin
- Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, China
| | - Tianjun Huang
- Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, China
| | - Youlin Li
- Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, China
| | - Chunjiang Zhu
- Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, China
| |
Collapse
|
32
|
Deng J, Wang X, Yan C, Huang Z, Luo H, Dai C, Huang X, Huang Y, Fu Q. Dietary purslane modulates gut microbiota and fecal metabolites in aging rats. Front Microbiol 2025; 16:1549853. [PMID: 40177479 PMCID: PMC11961954 DOI: 10.3389/fmicb.2025.1549853] [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: 12/22/2024] [Accepted: 02/27/2025] [Indexed: 04/05/2025] Open
Abstract
Introduction Portulaca oleracea L. (purslane) is a highly nutritious and edible wild vegetable beneficial to human health. However, its impacts on the structure of gut microbiota and fecal metabolites in aging individuals remain unclear. This study aims to clarify its potential mechanisms in aging-related gut health. Methods Naturally aged rats (18 months) were divided into two groups. One group was fed a maintenance chow, and the other was fed a mixture with 3.5% purslane for 15 weeks. Hematoxylin-eosin staining, gas chromatography-mass spectrometry, and 16S rDNA high-throughput sequencing were employed to explore the effects of purslane on the intestinal health of these rats. Results The fecal concentrations of acetic acid, propionic acid, butyric acid, valeric acid, caproic acid, and total short-chain fatty acids (SCFAs) were significantly increased in aging rats fed the purslane supplement. Purslane significantly reduced the relative abundance levels of Firmicutes and Fusobacteria, as well as the ratio of Firmicutes to Bacteroidetes. KEGG pathway analysis annotated 109 differential metabolites, which mainly affected metabolic pathways such as linoleic acid metabolism, arachidonic acid metabolism, primary bile acid biosynthesis, steroid biosynthesis, and steroid hormone biosynthesis. There was a strong correlation between Paracbacteroides, the Prevotella NK3B31_group, the Rikenella_RC9_gut_group, and SCFA levels. Aging rats consuming purslane had a more complete and healthy gut morphology than the control group. Discussion These results suggested that the maintenance of intestinal health by purslane in aging rats might be associated with the targeted regulation of gut microbiota and fecal metabolites.
Collapse
Affiliation(s)
- Jingwen Deng
- Affiliated Hospital of Jinggangshan University, Ji’an, China
- Jiangxi Province Key Laboratory of Organ Development and Epigenetics, Clinical Medical Research Center, Affiliated Hospital of Jinggangshan University, Medical Department of Jinggangshan University, Ji'an, China
| | - Xia Wang
- Department of Pathology, Ji'an Central People's Hospital, Ji'an, China
| | - Can Yan
- Affiliated Hospital of Jinggangshan University, Ji’an, China
- Jiangxi Province Key Laboratory of Organ Development and Epigenetics, Clinical Medical Research Center, Affiliated Hospital of Jinggangshan University, Medical Department of Jinggangshan University, Ji'an, China
| | - Zicheng Huang
- College of Mathematics and Physics, Xinjiang Agricultural University, Urumqi, China
| | - Hui Luo
- Jiangxi Province Key Laboratory of Organ Development and Epigenetics, Clinical Medical Research Center, Affiliated Hospital of Jinggangshan University, Medical Department of Jinggangshan University, Ji'an, China
| | - Caihua Dai
- Affiliated Hospital of Jinggangshan University, Ji’an, China
| | - Xiaoliu Huang
- Affiliated Hospital of Jinggangshan University, Ji’an, China
- Jiangxi Province Key Laboratory of Organ Development and Epigenetics, Clinical Medical Research Center, Affiliated Hospital of Jinggangshan University, Medical Department of Jinggangshan University, Ji'an, China
| | - Yushan Huang
- Center for Evidence Based Medical and Clinical Research, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Qiang Fu
- Affiliated Hospital of Jinggangshan University, Ji’an, China
- Jiangxi Province Key Laboratory of Organ Development and Epigenetics, Clinical Medical Research Center, Affiliated Hospital of Jinggangshan University, Medical Department of Jinggangshan University, Ji'an, China
| |
Collapse
|
33
|
Hyder N, Raza ML. Stress and the gut microbiota-brain axis. PROGRESS IN BRAIN RESEARCH 2025; 291:175-203. [PMID: 40222779 DOI: 10.1016/bs.pbr.2025.01.002] [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: 04/15/2025]
Abstract
The gut microbiota-brain axis is a complex system that links the bacteria in our gut with our brain, it plays a part in what way we respond to stress. This chapter explores how stress affects the types of bacteria in the gut and shows the two-way connection between them. Stress can change the bacteria in our gut, which can cause various problems related to stress, like depression, anxiety, and irritable bowel syndrome (IBS). Figuring out how these interactions may help us develop new treatments that focus on the connection between gut bacteria and the brain. This chapter looks at how gut bacteria could help identify stress-related problems. It also discusses the difficulties and possibilities of using this research in medical practice. In the end, the chapter talks about what comes next in this quickly changing area. It highlights how important it is to include research about the gut-brain connection in overall public health plans.
Collapse
Affiliation(s)
- Noorulain Hyder
- Department of Pharmacology, Faculty of Pharmacy, Hamdard University, Karachi, Pakistan; HEJ Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan.
| | - Muhammad Liaquat Raza
- Department of Infection Prevention & Control, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia; King Abdullah International Medical Research Center, Riyadh, Saudi Arabia; King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia.
| |
Collapse
|
34
|
Jiang Q, Wu L, Wang X, Gao Z, Liu X, Zhang W, Xue L, Yang J, Chen T, Chen Y, Wang B. Investigating Causal Links Between Gut Microbiota and Neurological Disorders via Genome-Wide Association Studies. Mol Neurobiol 2025:10.1007/s12035-025-04770-3. [PMID: 40075040 DOI: 10.1007/s12035-025-04770-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 02/10/2025] [Indexed: 03/14/2025]
Abstract
Many reports have highlighted the involvement of the gut microbiome in the occurrence, progression, and outcomes of neurological disorders. However, current reports are somewhat chaotic, especially concerning whether the gut microbiota has a causal effect on various neurological diseases. Furthermore, whether there is a common mechanism involving gut microbial communities in these neurological disorders has not to be revealed. In this study, we leveraged data from the largest-scale genome-wide association study (GWAS) by the MiBioGen consortium, which includes genetic and microbial composition data from 18,340 individuals spanning 24 cohorts. We utilized single-nucleotide polymorphisms (SNPs) associated with the gut microbiome as instrumental variables (IVs) in Mendelian randomization (MR) analyses. These IVs were rigorously selected based on their genome-wide and locus-wide significance to ensure robust causal inference. Our study established robust associations between specific gut microbiota and various neurological disorders using MR. We systematically depicted the bacteria with causal relationships in all diseases, covering the levels of phylum, class, order, family, and genus. We identified 34 bacterial species as significant risk or protective factors across disorders, including two main phylum levels such as Firmicutes (22 species) and Proteobacteria (8 species), as well as Bacteroidetes (2 species), Actinobacteria (1 species), and Verrucomicrobiota (1 species). At the family level of bacteria, we found that Lachnospiraceae and Ruminococcaceae are the most related to these 11 diseases and they may play different roles in the same disease.
Collapse
Affiliation(s)
- Qingchen Jiang
- Department of Immunology and Pathogenic Biology, College of Basic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, 76 Yanta West Road, 710061, China
| | - Laiqiang Wu
- College of Forensic Science, Key Laboratory of National Health Commission for Forensic Science, Xi'an Jiaotong University, Xi'an, China
| | - Xiaochen Wang
- College of Forensic Science, Key Laboratory of National Health Commission for Forensic Science, Xi'an Jiaotong University, Xi'an, China
| | - Zhe Gao
- School of Human Settlement and Civil Engineering, Xi'an Jiaotong University, Xi'an, China
| | - Xiaorui Liu
- College of Forensic Science, Key Laboratory of National Health Commission for Forensic Science, Xi'an Jiaotong University, Xi'an, China
| | - Wen Zhang
- Department of Pathology, Northwest Women'S and Children'S Hospital, Xi'an, China
| | - Li Xue
- Department of Clinical Laboratory, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Juan Yang
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Teng Chen
- College of Forensic Science, Key Laboratory of National Health Commission for Forensic Science, Xi'an Jiaotong University, Xi'an, China
| | - Yanjiong Chen
- Department of Immunology and Pathogenic Biology, College of Basic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, 76 Yanta West Road, 710061, China.
| | - Biao Wang
- Department of Immunology and Pathogenic Biology, College of Basic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, 76 Yanta West Road, 710061, China.
| |
Collapse
|
35
|
Fu J, Liang Z, Chen Z, Zhou Y, Xiong F, Liang Q, Gao H. Deciphering the Therapeutic Efficacy and Underlying Mechanisms of Dendrobium officinale Polysaccharides in the Intervention of Alzheimer's Disease Mice: Insights from Metabolomics and Microbiome. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:5635-5648. [PMID: 39536176 DOI: 10.1021/acs.jafc.4c07913] [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: 11/16/2024]
Abstract
As a traditional drug-food homologous plant, Dendrobium officinale is widely recognized for its nutritional and medicinal value. Specifically, D. officinale polysaccharide (DOP) has garnered attention as a potential prebiotic for its protective effects on gut microbiota and the nervous system. However, the underlying mechanism by which DOP improves cognitive dysfunction in Alzheimer's disease (AD) remains unclear. This study intends to elucidate the beneficial effects of DOP on AD mice from the perspectives of metabolomics and the intestinal microbiome. The results showed that DOP significantly ameliorated cognitive dysfunction, attenuated hippocampal neuronal damage and Aβ plaque deposition, and restored intestinal barrier integrity in AD mice. The antibiotic-cocktail-induced germ-free mouse model confirmed that the neuroprotective effect of DOP was dependent on gut microbiota. Further investigations demonstrated that DOP influenced the composition of gut microbiota and restored its diversity. Additionally, DOP reshaped metabolic profile disorders in AD mice and increased the short-chain fatty acids (SCFAs) content. Correlation analysis further highlighted that specific gut microbiota was associated with the metabolism of AD mice. In conclusion, this study sheds light on the positive impact of DOP in reshaping the gut microbiota and enhancing cognitive function, offering important perspectives for the possible advancement and utilization of DOP.
Collapse
Affiliation(s)
- Jun Fu
- Innovation Academy of Testing Technology, Scientific Research Center, Wenzhou Medical University, Wenzhou 325035, China
- Key Laboratory of Efficacy Evaluation of Traditional Chinese Medicine and Encephalopathy Research of Zhejiang Province, Wenzhou Medical University, Wenzhou 325035, China
| | - Zhaohan Liang
- Innovation Academy of Testing Technology, Scientific Research Center, Wenzhou Medical University, Wenzhou 325035, China
| | - Zihao Chen
- Innovation Academy of Testing Technology, Scientific Research Center, Wenzhou Medical University, Wenzhou 325035, China
| | - Yiyang Zhou
- Innovation Academy of Testing Technology, Scientific Research Center, Wenzhou Medical University, Wenzhou 325035, China
| | - Fen Xiong
- Innovation Academy of Testing Technology, Scientific Research Center, Wenzhou Medical University, Wenzhou 325035, China
| | - Qian Liang
- Innovation Academy of Testing Technology, Scientific Research Center, Wenzhou Medical University, Wenzhou 325035, China
- Key Laboratory of Efficacy Evaluation of Traditional Chinese Medicine and Encephalopathy Research of Zhejiang Province, Wenzhou Medical University, Wenzhou 325035, China
| | - Hongchang Gao
- Innovation Academy of Testing Technology, Scientific Research Center, Wenzhou Medical University, Wenzhou 325035, China
- Key Laboratory of Efficacy Evaluation of Traditional Chinese Medicine and Encephalopathy Research of Zhejiang Province, Wenzhou Medical University, Wenzhou 325035, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health); Institute of Metabonomics & Medical NMR, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| |
Collapse
|
36
|
Li J, Liu T, Xian M, Zhou K, Wei J. The Power of Exercise: Unlocking the Biological Mysteries of Peripheral-Central Crosstalk in Parkinson's Disease. J Adv Res 2025:S2090-1232(25)00143-2. [PMID: 40049515 DOI: 10.1016/j.jare.2025.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 01/06/2025] [Accepted: 03/01/2025] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND Exercise is a widely recognized non-pharmacological treatment for Parkinson's Disease (PD). The bidirectional regulation between the brain and peripheral organs has emerged as a promising area of research, with the mechanisms by which exercise impacts PD closely linked to the interplay between peripheral signals and the central nervous system. AIM OF REVIEW This review aims to summarize the mechanisms by which exercise influences peripheral-central crosstalk to improve PD, discuss the molecular processes mediating these interactions, elucidate the pathways through which exercise may modulate PD pathophysiology, and identify directions for future research. KEY SCIENTIFIC CONCEPTS OF REVIEW This review examines how exercise-induced cytokine release promotes neuroprotection in PD. It discusses how exercise can stimulate cytokine secretion through various pathways, including the gut-brain, muscle-brain, liver-brain, adipose-brain, and bone-brain axes, thereby alleviating PD symptoms. Additionally, the potential contributions of the heart-brain, lung-brain, and spleen-brain axes, as well as multi-axis crosstalk-such as the brain-gut-muscle and brain-gut-bone axes-are explored in the context of exercise therapy. The study highlights the need for further research into peripheral-central crosstalk and outlines future directions to address challenges in clinical PD therapy.
Collapse
Affiliation(s)
- Jingwen Li
- Institute for Sports and Brain Health, School of Physical Education, Henan University, Kaifeng, Henan, 475004, China
| | - Tingting Liu
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Meiyan Xian
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Ke Zhou
- Institute for Sports and Brain Health, School of Physical Education, Henan University, Kaifeng, Henan, 475004, China.
| | - Jianshe Wei
- Institute for Sports and Brain Health, School of Physical Education, Henan University, Kaifeng, Henan, 475004, China; Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China.
| |
Collapse
|
37
|
Tadese DA, Mwangi J, Luo L, Zhang H, Huang X, Michira BB, Zhou S, Kamau PM, Lu Q, Lai R. The microbiome's influence on obesity: mechanisms and therapeutic potential. SCIENCE CHINA. LIFE SCIENCES 2025; 68:657-672. [PMID: 39617855 DOI: 10.1007/s11427-024-2759-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 10/16/2024] [Indexed: 01/03/2025]
Abstract
In 2023, the World Obesity Atlas Federation concluded that more than 50% of the world's population would be overweight or obese within the next 12 years. At the heart of this epidemic lies the gut microbiota, a complex ecosystem that profoundly influences obesity-related metabolic health. Its multifaced role encompasses energy harvesting, inflammation, satiety signaling, gut barrier function, gut-brain communication, and adipose tissue homeostasis. Recognizing the complexities of the cross-talk between host physiology and gut microbiota is crucial for developing cutting-edge, microbiome-targeted therapies to address the global obesity crisis and its alarming health and economic repercussions. This narrative review analyzed the current state of knowledge, illuminating emerging research areas and their implications for leveraging gut microbial manipulations as therapeutic strategies to prevent and treat obesity and related disorders in humans. By elucidating the complex relationship between gut microflora and obesity, we aim to contribute to the growing body of knowledge underpinning this critical field, potentially paving the way for novel interventions to combat the worldwide obesity epidemic.
Collapse
Affiliation(s)
- Dawit Adisu Tadese
- Engineering Laboratory of Peptides of Chinese Academy of Sciences, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Key Laboratory of Genetic Evolution & Animal Models, Sino-African Joint Research Center, and New Cornerstone Science Laboratory, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China
- Kunming College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - James Mwangi
- Engineering Laboratory of Peptides of Chinese Academy of Sciences, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Key Laboratory of Genetic Evolution & Animal Models, Sino-African Joint Research Center, and New Cornerstone Science Laboratory, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China
- Kunming College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Lei Luo
- Engineering Laboratory of Peptides of Chinese Academy of Sciences, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Key Laboratory of Genetic Evolution & Animal Models, Sino-African Joint Research Center, and New Cornerstone Science Laboratory, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China
- Kunming College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Hao Zhang
- Engineering Laboratory of Peptides of Chinese Academy of Sciences, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Key Laboratory of Genetic Evolution & Animal Models, Sino-African Joint Research Center, and New Cornerstone Science Laboratory, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China
| | - Xiaoshan Huang
- Engineering Laboratory of Peptides of Chinese Academy of Sciences, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Key Laboratory of Genetic Evolution & Animal Models, Sino-African Joint Research Center, and New Cornerstone Science Laboratory, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China
- Kunming College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Brenda B Michira
- Engineering Laboratory of Peptides of Chinese Academy of Sciences, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Key Laboratory of Genetic Evolution & Animal Models, Sino-African Joint Research Center, and New Cornerstone Science Laboratory, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China
- Kunming College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Shengwen Zhou
- Engineering Laboratory of Peptides of Chinese Academy of Sciences, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Key Laboratory of Genetic Evolution & Animal Models, Sino-African Joint Research Center, and New Cornerstone Science Laboratory, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China
- Kunming College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Peter Muiruri Kamau
- Engineering Laboratory of Peptides of Chinese Academy of Sciences, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Key Laboratory of Genetic Evolution & Animal Models, Sino-African Joint Research Center, and New Cornerstone Science Laboratory, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China
- Kunming College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Qiumin Lu
- Engineering Laboratory of Peptides of Chinese Academy of Sciences, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Key Laboratory of Genetic Evolution & Animal Models, Sino-African Joint Research Center, and New Cornerstone Science Laboratory, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China
| | - Ren Lai
- Engineering Laboratory of Peptides of Chinese Academy of Sciences, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Key Laboratory of Genetic Evolution & Animal Models, Sino-African Joint Research Center, and New Cornerstone Science Laboratory, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China.
- Kunming College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
38
|
Li J, Lv J, Zhang Y, Zhou Z, Geng H, Zhou Y, Yang C, Feng N. Inverted U-shape association between urine equol levels and cancer: a national population-based cross-sectional study. Sci Rep 2025; 15:7114. [PMID: 40016447 PMCID: PMC11868629 DOI: 10.1038/s41598-025-91846-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 02/24/2025] [Indexed: 03/01/2025] Open
Abstract
Equol, a naturally occurring phytoestrogen derived from the fermentation of soy and soy-based products by gut bacteria, is recognized for its diverse health benefits. While there is speculation about its association with cancer prevention, the scientific community has yet to reach a consensus due to the variability in research findings. Our study aims to shed light on this topic by examining the correlation between urine equol concentrations and the cancer risk among the American population. The National Health and Nutrition Examination Survey (NHANES) is a national survey of U.S. civilians in which cancer participants are enrolled in a database by a sample questionnaire. This study included 2797 Americans aged 40 years and older in the NHANES database (2005-2010). The relationship between urine equol concentration and cancer was analysed using weighted logistic regression models, stratified analysis, smoothed curve fitting and threshold effect analysis were also performed. Among the 2797 participants in our study, 390 individuals received a cancer diagnosis. Our findings indicate a positive correlation between urine equol levels and the risk of cancer. Notably, individuals in the highest quartile of equol excretion exhibited a significantly elevated risk of cancer, with a 25.4% increase compared to those in the lowest quartile (POR = 1.254, 95% CI: 1.252, 1.256), after fully adjusting for confounders. Similar results were observed in other adjusted models. A non-linear relationship in the shape of an inverted U-shape can be observed by smoothed curve fitting, and the inflection point is 25.5. Urinary equol concentrations below 25.5 ng/ml were positively associated with cancer risk, while equol concentrations above 25.5 ng/ml showed a slight negative trend in cancer risk. However, further prospective studies are needed to provide more robust evidence and confirmed in large clinical trials.
Collapse
Affiliation(s)
- Jufa Li
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Jing Lv
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Yuwei Zhang
- Medical School of Nantong University, Nantong, China
- Department of Urology, Jiangnan University Medical Center, Wuxi, China
| | - Zhihao Zhou
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Haochen Geng
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Yuhua Zhou
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Chun Yang
- Wuxi School of Medicine, Jiangnan University, Wuxi, China.
- Department of Urology, Jiangnan University Medical Center, Wuxi, China.
| | - Ninghan Feng
- Wuxi School of Medicine, Jiangnan University, Wuxi, China.
- Medical School of Nantong University, Nantong, China.
- Department of Urology, Jiangnan University Medical Center, Wuxi, China.
| |
Collapse
|
39
|
Chen X, Wei J, Zhang L, Wang H, Zhang Y, Li Z, Wang X, Liu L, Zhang Y, Zhang T. Association between plasma short-chain fatty acids and inflammation in human immunodeficiency virus-associated neurocognitive disorder: a pilot study. Lipids Health Dis 2025; 24:66. [PMID: 39984934 PMCID: PMC11846350 DOI: 10.1186/s12944-025-02477-x] [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: 01/06/2025] [Accepted: 02/10/2025] [Indexed: 02/23/2025] Open
Abstract
BACKGROUND AND AIMS Short-chain fatty acids (SCFAs), key metabolites produced by gut microbiota, have neuroprotective effects in neurodegenerative diseases by modulating immune responses. However, their role in human immunodeficiency virus (HIV)-associated neurocognitive disorder (HAND) remains largely unexplored. METHODS We recruited HAND patients, HIV Control, and healthy controls (HC). Plasma SCFAs and SCFA-producing gut microbiota were quantified via gas chromatography-mass spectrometry and fecal metagenomic analysis. Inflammatory cytokine levels were measured using liquid chromatography. Receiver operating characteristic (ROC) curves were generated to evaluate the predictive accuracy of SCFAs for HAND. RESULTS Plasma SCFAs were significantly reduced in HAND patients, correlating with a decrease in SCFA-producing gut bacteria, such as Prevotella and its related species. Reduced SCFAs were positively correlated with pro-inflammatory cytokines and cognitive impairment, while being negatively correlated with anti-inflammatory cytokines. ROC curve analysis demonstrated that several SCFAs exhibited strong predictive accuracy for HAND status. CONCLUSIONS SCFAs may influence cognitive function by modulating inflammatory responses, and identifies plasma SCFAs as potential biomarkers and therapeutic targets for HAND. Further investigation is needed to delineate the mechanisms that SCFAs influence HAND pathology.
Collapse
Affiliation(s)
- Xue Chen
- Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory for HIV/AIDS Research, Beijing Youan Hospital, Capital Medical University, Beijing, China
- Beijing Youan Hospital, Beijing Institute of Hepatology, Capital Medical University, Beijing, China
| | - Jiaqi Wei
- Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory for HIV/AIDS Research, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Ling Zhang
- Beijing Youan Hospital, Beijing Institute of Hepatology, Capital Medical University, Beijing, China
- Department of Respiratory and Critical Care Medicine, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Hu Wang
- Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory for HIV/AIDS Research, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Yang Zhang
- Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory for HIV/AIDS Research, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Zhen Li
- Beijing Key Laboratory for HIV/AIDS Research, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Xia Wang
- Beijing Key Laboratory for HIV/AIDS Research, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Lifeng Liu
- Beijing Key Laboratory for HIV/AIDS Research, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Yulin Zhang
- Department of Respiratory and Critical Care Medicine, Beijing Youan Hospital, Capital Medical University, Beijing, China.
| | - Tong Zhang
- Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China.
- Beijing Key Laboratory for HIV/AIDS Research, Beijing Youan Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
40
|
Xie H, Zhang H, Zhou L, Chen J, Yao S, He Q, Li Z, Zhou Z. Fecal microbiota transplantation promotes functional recovery in mice with spinal cord injury by modulating the spinal cord microenvironment. J Transl Med 2025; 23:210. [PMID: 39979990 PMCID: PMC11843963 DOI: 10.1186/s12967-025-06232-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 02/11/2025] [Indexed: 02/22/2025] Open
Abstract
BACKGROUND spinal cord injury (SCI) disrupts the gut microbiota, worsening the injury's impact. Fecal microbiota transplantation (FMT) is increasingly recognized as a promising strategy to improve neural function post-SCI, yet its precise mechanisms are still far from clear. The present study aims to elucidate how FMT influences motor function recovery and its underlying mechanisms utilizing a SCI mouse model. METHODS Mice with SCI received FMT from healthy donors. We used 16 S rRNA amplicon sequencing to analyze the alterations of gut microbes. Pathological alterations in the spinal cord tissue, including neuronal survival, axonal regeneration, cell proliferation, and neuroinflammation, were assessed among experimental groups. Additionally, RNA sequencing (RNA-seq) was used to explore alterations in relevant signaling pathways. RESULTS Significant shifts in gut microbiota composition following SCI were observed through 16 S rRNA analysis. On day 7 post-SCI, the FMT group exhibited a significantly higher diversity of gut microbiota compared to the ABX group, with the composition in the FMT group more closely resembling that of healthy mice. FMT promoted neuronal survival and axonal regeneration, leading to notable improvements in motor function compared to control mice. Immunofluorescence staining showed increased neuronal survival, alleviated extracellular matrix (ECM) deposition, diminished glial scar formation, and reduced inflammation in FMT-treated mice. RNA-seq analysis indicated that FMT induced transcriptomic changes associated with material metabolism, ECM remodeling, and anti-inflammatory responses. CONCLUSIONS FMT restored gut microbiota balance in SCI mice, mitigated inflammation, and promoted ECM remodeling, establishing an optimal environment for neural recovery. These findings demonstrated that FMT may represent a valuable approach to enhance functional recovery following SCI.
Collapse
Affiliation(s)
- Huan Xie
- The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong Province, 510630, China
| | - Hui Zhang
- The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, Guangdong Province, 510317, China
| | - Liyi Zhou
- The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, Guangdong Province, 510317, China
| | - Junjie Chen
- The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, Guangdong Province, 510317, China
| | - Shun Yao
- The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, Guangdong Province, 510317, China
| | - Quanxin He
- The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, Guangdong Province, 510317, China
| | - Zhizhong Li
- The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong Province, 510630, China.
| | - Zhilai Zhou
- The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, Guangdong Province, 510317, China.
| |
Collapse
|
41
|
Xiao HL, Zhu H, Zeng TA, Xu F, Yu SH, Yang CJ. Potential similarities in gut microbiota composition between autism spectrum disorder and neurotypical siblings: Insights from a comprehensive meta-analysis. Neuroscience 2025; 567:172-181. [PMID: 39788315 DOI: 10.1016/j.neuroscience.2025.01.010] [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: 10/23/2024] [Revised: 12/22/2024] [Accepted: 01/06/2025] [Indexed: 01/12/2025]
Abstract
BACKGROUND Previous studies have explored the differences in gut microbiota (GM) between individuals with autism spectrum disorder (ASD) and neurotypical controls. However, factors such as diet, lifestyle, and environmental exposure influence GM, leading to significant variability, even among neurotypical individuals. Comparing the GM of ASD individuals with neurotypical siblings, who share similar genes and living conditions, may offer better insights into the GM mechanisms associated with ASD. Therefore, this study aims to analyze the GM composition in ASD by comparing it to that of neurotypical siblings, potentially identifying microbiota that influence ASD. METHODS We explored electronic databases up to July 2024, including EBSCOhost, PubMed, ScienceDirect, Web of Science, and Scopus. Meta-analysis using RevMan 5.4 assessed the relative abundance (RA) of gut bacteria from 8 phyla and 4 genera in ASD individuals and neurotypical siblings. RESULTS Eight studies were included, involving 248 people with ASD and 197 neurotypical siblings. Significant but unstable differences were observed in the RA of Bacteroidetes, Firmicutes, and Fusobacteria. No significant differences were found in the RA of Proteobacteria, Cyanobacteria, Actinobacteria, Verrucomicrobia, Tenericutes, or Bacteroides, Roseburia, Sutterella, Bifidobacterium. CONCLUSIONS GM composition in ASD individuals closely resembles that of neurotypical siblings, with only a few unstable differences. This suggests that other crucial bacteria or certain interacting environmental factors play a role. Further studies are needed to gather stronger evidence to uncover the differences in GM and their mechanisms in ASD people.
Collapse
Affiliation(s)
- Hong-Li Xiao
- Faculty of Education, East China Normal University, Shanghai, China
| | - Han Zhu
- Faculty of Education, East China Normal University, Shanghai, China
| | - Tong-Ao Zeng
- Faculty of Education, East China Normal University, Shanghai, China
| | - Fang Xu
- Faculty of Education, East China Normal University, Shanghai, China; Hangzhou Health Experimental School, Zhejiang, China
| | - Su-Hong Yu
- Faculty of Education, East China Normal University, Shanghai, China.
| | - Chang-Jiang Yang
- Faculty of Education, East China Normal University, Shanghai, China; China Research Institute of Care and Education of Infants and Young, ECNU, Shanghai, China.
| |
Collapse
|
42
|
Lu D, Ma X, Tao K, Lei H. Advancements in the Pathogenesis, Diagnosis, and Therapeutic Implications of Intestinal Bacteria. Curr Issues Mol Biol 2025; 47:106. [PMID: 39996827 PMCID: PMC11853859 DOI: 10.3390/cimb47020106] [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: 12/31/2024] [Revised: 01/28/2025] [Accepted: 02/06/2025] [Indexed: 02/26/2025] Open
Abstract
Intestinal bacteria form one of the most complex microbial communities in the human body, playing a crucial role in maintaining host health and contributing to the development of various diseases. Here, we provide a comprehensive overview of the composition and function of intestinal bacteria, the factors affecting their homeostasis, and their association and mechanisms with a range of diseases (e.g., inflammatory bowel diseases, colorectal cancer, metabolic diseases). Additionally, their advanced potential in disease diagnosis and treatment is highlighted. Therapies, such as chemotherapy, radiotherapy, and immunotherapy, are significantly impacted by intestinal bacteria, with research indicating that bacteria can enhance chemoimmunotherapy efficiency by affecting T cell recruitment and immune cell infiltration. Fecal microbiota transplantation has emerged as a promising option for treating recurrent Clostridium difficile infections and certain metabolic and neurological disorders. Gut bacteria-related serum metabolites serve as non-invasive indicators for diagnosing CRC, while fecal immunochemical tests offer promising applications in CRC screening. Future research is needed to better understand the causal relationships between intestinal bacteria and diseases, develop more precise diagnostic tools, and evaluate the effectiveness and safety of microbiome-targeted therapies in clinical treatment. This study provides deeper insights into the role of intestinal bacteria in human health and disease, providing a scientific basis for innovative therapeutic strategies that have the potential to transform the landscape of healthcare.
Collapse
Affiliation(s)
| | | | - Kaixiong Tao
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (D.L.); (X.M.)
| | - Hongwei Lei
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (D.L.); (X.M.)
| |
Collapse
|
43
|
Liu C, Lu Q, Xi Q, Xiao S, Du J, Qin R, Wang J, Xu B, Han X, Zhou K, Tao S, Lv H, Jiang Y, Jiang T, Ye K, Jin G, Ma H, Xia Y, Shen H, Liu X, Lin Y, Hu Z. Varying Bifidobacterium species in the maternal-infant gut microbiota correlate with distinct early neurodevelopmental outcomes. J Genet Genomics 2025:S1673-8527(25)00030-X. [PMID: 39923844 DOI: 10.1016/j.jgg.2025.01.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Accepted: 01/19/2025] [Indexed: 02/11/2025]
Abstract
The impact of mother-infant microbiota on neurodevelopment is an area of interest, but longitudinal studies are scarce. Using a cohort of 520 families from the Jiangsu birth cohort in China, we reveal that the maternal gut microbiota during early pregnancy play a substantial role, accounting for 3.34% of the variance in offspring neurodevelopmental scores. This contribution is notably higher than the 1.24% attributed to the infants' own microbiota at 1 year of age, underscoring the significant influence of maternal gut health on early child development. Remarkably, an elevation in maternal Bifidobacterium pseudocatenulatum is linked to decreased cognitive scores, whereas an enrichment of Bifidobacterium longum at 1 year of age is associated with higher cognitive scores. Furthermore, we find that maternal B. pseudocatenulatum is linked to the heterolactic fermentation metabolic pathway, while infant B. longum is associated with the Bifidobacterium shunt pathway. In summary, our analysis implies that maternal and infant gut microbiota play a distinct role in neurodevelopment, suggesting potential strategies for improving neurodevelopmental outcomes during early pregnancy or infant development by targeting gut microbiota composition.
Collapse
Affiliation(s)
- Cong Liu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China; Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Qun Lu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China; Department of Maternal, Child and Adolescent Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China; Department of Prenatal Diagnosis, Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing, Jiangsu 210004, China
| | - Qi Xi
- Department of Obstetrics, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu 215002, China
| | - Shuxin Xiao
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China; Department of Maternal, Child and Adolescent Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Jiangbo Du
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China; Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China; State Key Laboratory of Reproductive Medicine and Offspring Health (Suzhou Centre), The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu 215002, China
| | - Rui Qin
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China; Scientific Research and Education Department, Changzhou Maternity and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou, Jiangsu 213000, China
| | - Jinghan Wang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China; Department of Maternal, Child and Adolescent Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Bo Xu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China; Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Xiumei Han
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China; Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Kun Zhou
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China; Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Shiyao Tao
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Hong Lv
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China; Department of Maternal, Child and Adolescent Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China; State Key Laboratory of Reproductive Medicine and Offspring Health (Suzhou Centre), The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu 215002, China
| | - Yangqian Jiang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China; Department of Maternal, Child and Adolescent Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Tao Jiang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China; Department of Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Kan Ye
- Department of Child Health Care, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu 215002, China
| | - Guangfu Jin
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China; Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China; State Key Laboratory of Reproductive Medicine and Offspring Health (Suzhou Centre), The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu 215002, China
| | - Hongxia Ma
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China; Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China; State Key Laboratory of Reproductive Medicine and Offspring Health (Suzhou Centre), The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu 215002, China
| | - Yankai Xia
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China; Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Hongbing Shen
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China; Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China; State Key Laboratory of Reproductive Medicine and Offspring Health (Suzhou Centre), The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu 215002, China
| | - Xingyin Liu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China.
| | - Yuan Lin
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China; Department of Maternal, Child and Adolescent Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China; State Key Laboratory of Reproductive Medicine and Offspring Health (Suzhou Centre), The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu 215002, China.
| | - Zhibin Hu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China; Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China; State Key Laboratory of Reproductive Medicine and Offspring Health (Suzhou Centre), The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu 215002, China.
| |
Collapse
|
44
|
Li Y, Feng J, Ding G, Deng L, He Y, Zhang Q, Wang J, Chen X. The possible effects of chili peppers on ADHD in relation to the gut microbiota. Front Nutr 2025; 12:1551650. [PMID: 39968396 PMCID: PMC11832391 DOI: 10.3389/fnut.2025.1551650] [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: 12/26/2024] [Accepted: 01/22/2025] [Indexed: 02/20/2025] Open
Abstract
Attention deficit hyperactivity disorder (ADHD) is a common neurodevelopmental disorder, which is characterized by inattention, impulsivity and hyperactivity. Although the etiology and pathogenesis of ADHD are not fully understood, existing studies have shown that it may be related to genetic factors, environmental factors, abnormal brain development, and psychosocial factors. In recent years, with the concept of microbioa-gut-brain axis (MGBA), more and more studies have begun to pay attention to the effect of gut microbiota on ADHD. Dietary structure can significantly change the diversity and abundance of gut microbiota. Therefore, dietary supplements or food additives to regulate gut microbiota have become one of the potential ways to treat ADHD. Peppers, as an important dietary component, have potential value in regulating gut microbiota. Among them, capsaicin (8-methyl N-vanillyl-6-noneamide, CAP), as a key active component of peppers, has been shown to have potential therapeutic effects on central nervous system (CNS) diseases such as Parkinson's disease, epilepsy, and depression. In addition, much attention has been paid to the beneficial effects of CAP on gut microbiota. Chili peppers contain not only CAP, but also rich in vitamin C and fatty acids, all of which may ameliorate ADHD by modulating the gut microbiota. This finding not only provides a potential treatment for ADHD, but also provides a new perspective to expand the research and clinical treatment of ADHD pathogenesis. Although current research on the potential therapeutic effects of chili peppers on ADHD is still at an early stage and requires further verification through larger-scale and more rigorous controlled studies, its potential clinical value cannot be ignored.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Xia Chen
- Department of Pediatrics, Child and Adolescent Psychiatric Center of Jiangbei Campus, The First Affiliated Hospital of Army Medical University (Army 958th Hospital), Chongqing, China
| |
Collapse
|
45
|
Faraji N, Payami B, Ebadpour N, Gorji A. Vagus nerve stimulation and gut microbiota interactions: A novel therapeutic avenue for neuropsychiatric disorders. Neurosci Biobehav Rev 2025; 169:105990. [PMID: 39716559 DOI: 10.1016/j.neubiorev.2024.105990] [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: 11/18/2024] [Accepted: 12/19/2024] [Indexed: 12/25/2024]
Abstract
The rising prevalence of treatment-resistant neuropsychiatric disorders underscores the need for innovative and effective treatment strategies. The gut microbiota (GM) plays a pivotal role in the progression of these diseases, influencing the brain and mental health through the gut-brain axis (GBA). The vagus nerve plays a significant role in the GBA, making it a key area of focus for potential novel therapeutic interventions. Vagus nerve stimulation (VNS) was introduced and approved as a treatment for refractory forms of some neuropsychological disorders, such as depression and epilepsy. Considering its impact on several brain regions that play a vital part in mood, motivation, affection, and cognitive function, the VNS has shown significant therapeutic potential for treating a variety of neuropsychiatric disorders. Using VNS to target the bidirectional communication pathways linking the GM and the VN could present an exciting and novel approach to treating neuropsychological disorders. Imbalances in the GM, such as dysbiosis, can impair the communication pathways between the gut and the brain, contributing to the development of neuropsychological disorders. VNS shows potential for modulating these interconnected systems, helping to restore balance. Interestingly, the composition of the GM may also influence the effectiveness of VNS, as it has the potential to modify the brain's response to this therapeutic approach. This study provides a comprehensive analysis of a relatively unexplored but noteworthy interaction between VNS and GM in the treatment of neuropsychiatric disorders. In addition, we discussed the mechanisms, therapeutic potential, and clinical implications of VNS on the GBA across neuropsychiatric disorders.
Collapse
Affiliation(s)
- Navid Faraji
- Student research committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Bahareh Payami
- Student research committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Negar Ebadpour
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali Gorji
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Epilepsy Research Center, Department of Neurosurgery, Münster University, Germany; Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran.
| |
Collapse
|
46
|
Bruni O, Breda M, Mammarella V, Mogavero MP, Ferri R. Sleep and circadian disturbances in children with neurodevelopmental disorders. Nat Rev Neurol 2025; 21:103-120. [PMID: 39779841 DOI: 10.1038/s41582-024-01052-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/19/2024] [Indexed: 01/11/2025]
Abstract
Sleep is essential for brain development and overall health, particularly in children with neurodevelopmental disorders (NDDs). Sleep disruptions can considerably impact brain structure and function, leading to dysfunction of neurotransmitter systems, metabolism, hormonal balance and inflammatory processes, potentially contributing to the pathophysiology of NDDs. This Review examines the prevalence, types and mechanisms of sleep disturbances in children with NDDs, including autism spectrum disorder, attention-deficit hyperactivity disorder and various genetic syndromes. Common sleep disorders in these populations include insomnia, hypersomnia, circadian rhythm disorders, sleep-related breathing disorders and parasomnias, with underlying factors often involving genetic, neurobiological, environmental and neurophysiological influences. Sleep problems such as insomnia, night awakenings and sleep fragmentation are closely linked to both internalizing symptoms such as anxiety and depression, and externalizing behaviours such as hyperactivity and aggression. Assessment of sleep in children with NDDs presents unique challenges owing to communication difficulties, comorbid conditions and altered sensory processing. The Review underscores the importance of further research to unravel the complex interactions between sleep and neurodevelopment, advocating for longitudinal studies and the identification of predictive biomarkers. Understanding and addressing sleep disturbances in NDDs is crucial for improving developmental outcomes and the overall quality of life for affected individuals and their families.
Collapse
Affiliation(s)
- Oliviero Bruni
- Department of Developmental and Social Psychology, Sapienza University of Rome, Rome, Italy.
| | - Maria Breda
- Department of Psychology, Sapienza University of Rome, Rome, Italy
| | | | - Maria Paola Mogavero
- Vita-Salute San Raffaele University, Milan, Italy
- Sleep Disorders Centre, Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Raffaele Ferri
- Sleep Research Centre, Oasi Research Institute - IRCCS, Troina, Italy
| |
Collapse
|
47
|
Tomaszek N, Urbaniak AD, Bałdyga D, Chwesiuk K, Modzelewski S, Waszkiewicz N. Unraveling the Connections: Eating Issues, Microbiome, and Gastrointestinal Symptoms in Autism Spectrum Disorder. Nutrients 2025; 17:486. [PMID: 39940343 PMCID: PMC11819948 DOI: 10.3390/nu17030486] [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: 12/16/2024] [Revised: 01/27/2025] [Accepted: 01/28/2025] [Indexed: 02/16/2025] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental condition characterized by challenges in social communication, restricted interests, and repetitive behaviors. It is also associated with a high prevalence of eating disorders, gastrointestinal (GI) symptoms, and alterations in gut microbiota composition. One of the most pressing concerns is food selectivity. Various eating disorders, such as food neophobia, avoidant/restrictive food intake disorder (ARFID), specific dietary patterns, and poor-quality diets, are commonly observed in this population, often leading to nutrient deficiencies. Additionally, gastrointestinal problems in children with ASD are linked to imbalances in gut microbiota and immune system dysregulation. The aim of this narrative review is to identify previous associations between the gut-brain axis and gastrointestinal problems in ASD. We discuss the impact of the "microbiome-gut-brain axis", a bidirectional connection between gut microbiota and brain function, on the development and symptoms of ASD. In gastrointestinal problems associated with ASD, a 'vicious cycle' may play a significant role: ASD symptoms contribute to the prevalence of ARFID, which in turn leads to microbiota degradation, ultimately worsening ASD symptoms. Current data suggest a link between gastrointestinal problems in ASD and the microbiota, but the amount of evidence is limited. Further research is needed, targeting the correlation of a patient's microbiota status, dietary habits, and disease course.
Collapse
Affiliation(s)
| | | | | | | | - Stefan Modzelewski
- Department of Psychiatry, Medical University of Bialystok, pl. Wołodyjowskiego 2, 15-272 Białystok, Poland; (N.T.); (A.D.U.); (D.B.); (K.C.); (N.W.)
| | | |
Collapse
|
48
|
Beltrán-Velasco AI, Clemente-Suárez VJ. Harnessing Gut Microbiota for Biomimetic Innovations in Health and Biotechnology. Biomimetics (Basel) 2025; 10:73. [PMID: 39997096 PMCID: PMC11852373 DOI: 10.3390/biomimetics10020073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Revised: 01/20/2025] [Accepted: 01/23/2025] [Indexed: 02/26/2025] Open
Abstract
The gut microbiota is a complex and dynamic ecosystem that plays a fundamental role in human health by regulating immunity, metabolism, and the gut-brain axis. Beyond its critical physiological functions, it has emerged as a rich source of inspiration for biomimetic innovations in healthcare and biotechnology. This review explores the transformative potential of microbiota-based biomimetics, focusing on key biological mechanisms such as resilience, self-regulation, and quorum sensing. These mechanisms have inspired the development of innovative applications, including personalized probiotics, synbiotics, artificial microbiomes, bioinspired biosensors, and bioremediation systems. Such technologies aim to emulate and optimize the intricate functions of microbial ecosystems, addressing challenges in healthcare and environmental sustainability. The integration of advanced technologies, such as artificial intelligence, bioengineering, and multi-omics approaches, has further accelerated the potential of microbiota biomimetics. These tools enable the development of precision therapies tailored to individual microbiota profiles, enhance the efficacy of diagnostic systems, and facilitate the design of environmentally sustainable solutions, such as waste-to-energy systems and bioremediation platforms. Emerging areas of innovation, including gut-on-chip models and synthetic biology, offer unprecedented opportunities for studying and applying microbiota principles in controlled environments. Despite these advancements, challenges remain. The replication of microbial complexity in artificial environments, ethical concerns regarding genetically engineered microorganisms, and equitable access to advanced therapies are critical hurdles that must be addressed. This review underscores the importance of interdisciplinary collaboration and public awareness in overcoming these barriers and ensuring the responsible development of microbiota-based solutions. By leveraging the principles of microbial ecosystems, microbiota biomimetics represents a promising frontier in healthcare and sustainability. This approach has the potential to revolutionize therapeutic strategies, redefine diagnostic tools, and address global challenges, paving the way for a more personalized, efficient, and sustainable future in medicine and biotechnology.
Collapse
Affiliation(s)
- Ana Isabel Beltrán-Velasco
- NBC Group, Psychology Department, School of Life and Nature Sciences, Nebrija University, 28248 Madrid, Spain
| | - Vicente Javier Clemente-Suárez
- Faculty of Sports Sciences, Universidad Europea de Madrid, Tajo Street, s/n, 28670 Madrid, Spain;
- Grupo de Investigación en Cultura, Educación y Sociedad, Universidad de la Costa, Barranquilla 080002, Colombia
| |
Collapse
|
49
|
Boonchooduang N, Louthrenoo O, Likhitweerawong N, Kunasol C, Thonusin C, Sriwichaiin S, Nawara W, Chattipakorn N, Chattipakorn SC. Impact of psychostimulants on microbiota and short-chain fatty acids alterations in children with attention-deficit/hyperactivity disorder. Sci Rep 2025; 15:3034. [PMID: 39856212 PMCID: PMC11759945 DOI: 10.1038/s41598-025-87546-y] [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: 09/18/2024] [Accepted: 01/20/2025] [Indexed: 01/27/2025] Open
Abstract
Attention-deficit/hyperactivity disorder (ADHD), a common neurodevelopmental disorder in children, is associated with alterations in gut microbiota and short-chain fatty acids (SCFAs), which are metabolites influencing the gut-brain axis. Evidence suggests that psychostimulant medications, widely used to manage ADHD symptoms, may also impact gut microbiota composition and SCFA levels. This study explores these potential effects by examining gut microbiota profiles and SCFA concentrations in unmedicated and medicated children with ADHD, compared to healthy controls. Fecal samples from 30 children aged 6-12 years (10 unmedicated ADHD, 10 medicated ADHD, and 10 healthy controls) were analyzed using 16 S rRNA sequencing and targeted metabolomics. Unmedicated ADHD children show distinct gut microbiota profiles, with lower level of Tyzzerella, Prevotellaceae, and Coriobacteriaceae, compared to controls. Notably, propionic acid levels were negatively associated with ADHD symptom severity, suggesting a potential biomarker role. Medicated ADHD children showed lower gut microbial diversity, unique taxa, and lower SCFA levels, compared to unmedicated children with ADHD. These findings suggest that gut microbiota and SCFAs may be linked to ADHD symptomatology, underscoring the importance of gut-brain interactions in ADHD. This study highlights the potential of gut health monitoring as part of future ADHD management strategies.
Collapse
Affiliation(s)
- Nonglak Boonchooduang
- Division of Developmental and Behavioral Pediatrics, Department of Pediatrics, Faculty of Medicine, Chiang Mai University, 50200, Chiang Mai, Thailand
| | - Orawan Louthrenoo
- Division of Developmental and Behavioral Pediatrics, Department of Pediatrics, Faculty of Medicine, Chiang Mai University, 50200, Chiang Mai, Thailand
| | - Narueporn Likhitweerawong
- Division of Developmental and Behavioral Pediatrics, Department of Pediatrics, Faculty of Medicine, Chiang Mai University, 50200, Chiang Mai, Thailand
| | - Chanon Kunasol
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, 50200, Chiang Mai, Thailand
| | - Chanisa Thonusin
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, 50200, Chiang Mai, Thailand
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, 50200, Chiang Mai, Thailand
| | - Sirawit Sriwichaiin
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, 50200, Chiang Mai, Thailand
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, 50200, Chiang Mai, Thailand
| | - Wichwara Nawara
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, 50200, Chiang Mai, Thailand
| | - Nipon Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, 50200, Chiang Mai, Thailand
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, 50200, Chiang Mai, Thailand
- The Academy of Science, The Royal Society of Thailand, Bangkok, Thailand
| | - Siriporn C Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand.
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, 50200, Chiang Mai, Thailand.
- Department of Oral Biology and Diagnostic Sciences, Faculty of Dentistry, Chiang Mai University, Chiang Mai, Chiang Mai, 50200, Thailand.
| |
Collapse
|
50
|
Li T, Zhao Z, Peng M, Zhang L, Wang C, Luo F, Zeng M, Sun K, Fang Z, Luo Y, Xie Y, Lv C, Wang J, Huang JD, Zhou H, Sun H. Multi-omics analysis reveals the interplay between intratumoral bacteria and glioma. mSystems 2025; 10:e0045724. [PMID: 39660865 PMCID: PMC11748541 DOI: 10.1128/msystems.00457-24] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 10/29/2024] [Indexed: 12/12/2024] Open
Abstract
Emerging evidence highlights the potential impact of intratumoral microbiota on cancer. However, the microbial composition and function in glioma remains elusive. Consequently, our study aimed to investigate the microbial community composition in glioma tissues and elucidate its role in glioma development. We parallelly performed microbial profiling, transcriptome sequencing, and metabolomics detection on tumor and adjacent normal brain tissues obtained from 50 glioma patients. We employed immunohistochemistry, multicolor immunofluorescence, and fluorescence in situ hybridization (FISH) staining to observe the presence and location of bacteria. Furthermore, an animal model was employed to validate the impact of key bacteria on glioma development. Six genera were found to be significantly enriched in glioma tissues compared to adjacent normal brain tissues, including Fusobacterium, Longibaculum, Intestinimonas, Pasteurella, Limosilactobacillus, and Arthrobacter. Both bacterial RNA and lipopolysaccharides (LPS) were observed in glioma tissues. Integrated microbiomics, transcriptomics, and metabolomics revealed that genes associated with intratumoral microbes were enriched in multiple synapse-associated pathways and that metabolites associated with intratumoral microbes were (R)-N-methylsalsolinol, N-acetylaspartylglutamic acid, and N-acetyl-l-aspartic acid. Further mediation analysis suggested that the intratumoral microbiome may affect the expression of neuron-related genes through bacteria-associated metabolites. In addition, both in vivo and in vitro models of glioma show that Fusobacterium nucleatum promotes glioma proliferation and upregulates CCL2, CXCL1, and CXCL2 levels. Our findings shed light on the intricate interplay between intratumoral bacteria and glioma. IMPORTANCE Our study adopted a multi-omics approach to unravel the impact of intratumoral microbes on neuron-related gene expression through bacteria-associated metabolites. Importantly, we found bacterial RNA and LPS signals within glioma tissues, which were traditionally considered sterile. We identified key microbiota within glioma tissues, including Fusobacterium nucleatum (Fn). Through in vivo and in vitro experiments, we identified the crucial role of Fn in promoting glioma progression, suggesting that Fn could be a potential diagnostic and therapeutic target for glioma patients. These findings offer valuable insights into the intricate interplay between intratumoral bacteria and glioma, offering novel inspiration to the realm of glioma biology.
Collapse
Affiliation(s)
- Ting Li
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Guangdong Provincial Clinical Research Center for Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Zhanyi Zhao
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Guangdong Provincial Clinical Research Center for Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Meichang Peng
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Guangdong Provincial Clinical Research Center for Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Lu Zhang
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Guangdong Provincial Clinical Research Center for Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Cheng Wang
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Guangdong Provincial Clinical Research Center for Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Feiyang Luo
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Guangdong Provincial Clinical Research Center for Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Meiqin Zeng
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Guangdong Provincial Clinical Research Center for Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Kaijian Sun
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Guangdong Provincial Clinical Research Center for Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Zhencheng Fang
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Guangdong Provincial Clinical Research Center for Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yunhao Luo
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Guangdong Provincial Clinical Research Center for Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yugu Xie
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Guangdong Provincial Clinical Research Center for Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Cui Lv
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Guangdong Provincial Clinical Research Center for Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jiaxuan Wang
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Guangdong Provincial Clinical Research Center for Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jian-Dong Huang
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Guangdong Provincial Clinical Research Center for Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong Special Administrative Region, China
- Chinese Academy of Sciences (CAS) Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Clinical Oncology Center, Shenzhen Key Laboratory for Cancer Metastasis and Personalized Therapy, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
- Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Hongwei Zhou
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Guangdong Provincial Clinical Research Center for Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Haitao Sun
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Guangdong Provincial Clinical Research Center for Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Southern Medical University, Guangzhou, China
| |
Collapse
|