1
|
Su Y, Liu N, Wang P, Shang C, Sun R, Ma J, Li Z, Ma H, Sun Y, Zhang Z, Song J, Xie Z, Xu J, Zhang Z. Proteomic analysis and experimental validation reveal the blood-brain barrier protective of Huanshaodan in the treatment of SAMP8 mouse model of Alzheimer's disease. Chin Med 2024; 19:137. [PMID: 39369234 PMCID: PMC11456246 DOI: 10.1186/s13020-024-01016-7] [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: 11/28/2023] [Accepted: 09/26/2024] [Indexed: 10/07/2024] Open
Abstract
BACKGROUND Huanshaodan (HSD) is a Chinese Herbal Compound which has a definite clinical effect on Alzheimer's disease (AD), however, the underlying mechanism remains unclear. The aim of this study is to preliminarily reveal the mechanism of HSD in the treatment of AD model of SAMP8 mice. METHODS Chemical composition of HSD and its drug-containing serum were identified by Q-Orbitrap high resolution liquid mass spectrometry. Six-month-old SAMP8 mice were treated with HSD and Donepezil hydrochloride by gavage for 2 months, and Wogonin for 28 days. Behavioral test was performed to test the learning and memory ability of mice. Immunofluorescence (IF) or Western-blot methods were used to detect the levels of pSer404-tau and β-amyloid (Aβ) in the brain of mice. Hematoxylin-eosin (H&E) staining and Transmission electron microscopy (TEM) assay was applied to observe the pathological changes of neurons. Proteomic technology was carried out to analyze and identify the protein network of HSD interventions in AD. Then the pathological process of the revealed AD-related differential proteins was investigated by IF, Q-PCR, Western-blot, Fluorescence in situ hybridization (FISH) and 16S rRNA sequencing methods. RESULTS The results showed that HSD and Wogonin, one of the components in its drug-containing serum, can effectively improve the cognitive impairments of SAMP8 mice, protect hippocampal neurons and synapses, and reduce the expression of pSer404-tau and Aβ. HSD and Wogonin reduced the levels of fibrinogen β chain (FGB) and γ chain (FGG), the potential therapeutic targets revealed by proteomics analysis, reduced the colocalization of FGB and FGG with Aβ, ionized calcium binding adaptor molecule 1 (Iba-1), glial fibrillary acidic protein (GFAP), increased level of and myelin basic protein (MBP). Meanwhile, HSD and Wogonin increased ZO-1 and Occludin levels, improved brain microvascular injury, and reduced levels of bacteria/bacterial DNA and lipopolysaccharide (LPS) in the brain of mice. In addition, 16S rRNA sequencing indicated that HSD regulated the structure of intestinal microbiota of mice. CONCLUSION The effects of HSD on AD may be achieved by inhibiting the levels of fibrinogen and the interactions on glia cells in the brain, and by modulating the structure of intestinal microbiota and improving the blood-brain barrier function.
Collapse
Affiliation(s)
- Yunfang Su
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province; Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, No. 156, Jinshuidong Road, Zhengzhou, 450046, China
- The First Affiliated Hospital of Henan University of Chinese Medicine, No. 19, Renmin Road, Zhengzhou, 450046, China
| | - Ningning Liu
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province; Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, No. 156, Jinshuidong Road, Zhengzhou, 450046, China
| | - Pan Wang
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province; Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, No. 156, Jinshuidong Road, Zhengzhou, 450046, China
| | - Congcong Shang
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province; Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, No. 156, Jinshuidong Road, Zhengzhou, 450046, China
| | - Ruiqin Sun
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province; Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, No. 156, Jinshuidong Road, Zhengzhou, 450046, China
| | - Jinlian Ma
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province; Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, No. 156, Jinshuidong Road, Zhengzhou, 450046, China
| | - Zhonghua Li
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province; Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, No. 156, Jinshuidong Road, Zhengzhou, 450046, China
| | - Huifen Ma
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province; Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, No. 156, Jinshuidong Road, Zhengzhou, 450046, China
| | - Yiran Sun
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province; Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, No. 156, Jinshuidong Road, Zhengzhou, 450046, China
| | - Zijuan Zhang
- School of Basic Medical Sciences, Henan University of Chinese Medicine, No. 156, Jinshuidong Road, Zhengzhou, 450046, China
| | - Junying Song
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province; Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, No. 156, Jinshuidong Road, Zhengzhou, 450046, China
| | - Zhishen Xie
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province; Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, No. 156, Jinshuidong Road, Zhengzhou, 450046, China.
| | - Jiangyan Xu
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province; Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, No. 156, Jinshuidong Road, Zhengzhou, 450046, China.
| | - Zhenqiang Zhang
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province; Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, No. 156, Jinshuidong Road, Zhengzhou, 450046, China.
| |
Collapse
|
2
|
Li S, Zhao L, Xiao J, Guo Y, Fu R, Zhang Y, Xu S. The gut microbiome: an important role in neurodegenerative diseases and their therapeutic advances. Mol Cell Biochem 2024; 479:2217-2243. [PMID: 37787835 DOI: 10.1007/s11010-023-04853-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 09/06/2023] [Indexed: 10/04/2023]
Abstract
There are complex interactions between the gut and the brain. With increasing research on the relationship between gut microbiota and brain function, accumulated clinical and preclinical evidence suggests that gut microbiota is intimately involved in the pathogenesis of neurodegenerative diseases (NDs). Increasingly studies are beginning to focus on the association between gut microbiota and central nervous system (CNS) degenerative pathologies to find potential therapies for these refractory diseases. In this review, we summarize the changes in the gut microbiota in Alzheimer's disease, Parkinson's disease, multiple sclerosis, and amyotrophic lateral sclerosis and contribute to our understanding of the function of the gut microbiota in NDs and its possible involvement in the pathogenesis. We subsequently discuss therapeutic approaches targeting gut microbial abnormalities in these diseases, including antibiotics, diet, probiotics, and fecal microbiota transplantation (FMT). Furthermore, we summarize some completed and ongoing clinical trials of interventions with gut microbes for NDs, which may provide new ideas for studying NDs.
Collapse
Affiliation(s)
- Songlin Li
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Linna Zhao
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, China
| | - Jie Xiao
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yuying Guo
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, China
| | - Rong Fu
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yunsha Zhang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Shixin Xu
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China.
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.
- Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, China.
| |
Collapse
|
3
|
Cocean AM, Vodnar DC. Exploring the gut-brain Axis: Potential therapeutic impact of Psychobiotics on mental health. Prog Neuropsychopharmacol Biol Psychiatry 2024; 134:111073. [PMID: 38914414 DOI: 10.1016/j.pnpbp.2024.111073] [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: 01/11/2024] [Revised: 04/18/2024] [Accepted: 06/17/2024] [Indexed: 06/26/2024]
Abstract
One of the most challenging and controversial issues in microbiome research is related to gut microbial metabolism and neuropsychological disorders. Psychobiotics affect human behavior and central nervous system processes via the gut-brain axis, involving neuronal, immune, and metabolic pathways. They have therapeutic potential in the treatment of several neurodegenerative and neurodevelopmental disorders such as depression, anxiety, autism, attention deficit hyperactivity disorder, Alzheimer's disease, Parkinson's disease, schizophrenia, Huntington's disease, anorexia nervosa, and multiple sclerosis. However, the mechanisms underlying the interaction between psychobiotics and the abovementioned diseases need further exploration. This review focuses on the relationship between gut microbiota and its impact on neurological and neurodegenerative disorders, examining the potential of psychobiotics as a preventive and therapeutic approach, summarising recent research on the gut-brain axis and the potential beneficial effects of psychobiotics, highlighting the need for further research and investigation in this area.
Collapse
Affiliation(s)
- Ana-Maria Cocean
- Department of Food Science and Technology, Life Science Institute, University of Agricultural Sciences and Veterinary Medicine, Calea Mănăștur 3-5, Cluj-Napoca, Romania.
| | - Dan Cristian Vodnar
- Department of Food Science and Technology, Life Science Institute, University of Agricultural Sciences and Veterinary Medicine, Calea Mănăștur 3-5, Cluj-Napoca, Romania.
| |
Collapse
|
4
|
Soraci L, Corsonello A, Paparazzo E, Montesanto A, Piacenza F, Olivieri F, Gambuzza ME, Savedra EV, Marino S, Lattanzio F, Biscetti L. Neuroinflammaging: A Tight Line Between Normal Aging and Age-Related Neurodegenerative Disorders. Aging Dis 2024; 15:1726-1747. [PMID: 38300639 PMCID: PMC11272206 DOI: 10.14336/ad.2023.1001] [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: 08/12/2023] [Accepted: 10/01/2023] [Indexed: 02/02/2024] Open
Abstract
Aging in the healthy brain is characterized by a low-grade, chronic, and sterile inflammatory process known as neuroinflammaging. This condition, mainly consisting in an up-regulation of the inflammatory response at the brain level, contributes to the pathogenesis of age-related neurodegenerative disorders. Development of this proinflammatory state involves the interaction between genetic and environmental factors, able to induce age-related epigenetic modifications. Indeed, the exposure to environmental compounds, drugs, and infections, can contribute to epigenetic modifications of DNA methylome, histone fold proteins, and nucleosome positioning, leading to epigenetic modulation of neuroinflammatory responses. Furthermore, some epigenetic modifiers, which combine and interact during the life course, can contribute to modeling of epigenome dynamics to sustain, or dampen the neuroinflammatory phenotype. The aim of this review is to summarize current knowledge about neuroinflammaging with a particular focus on epigenetic mechanisms underlying the onset and progression of neuroinflammatory cascades in the central nervous system; furthermore, we describe some diagnostic biomarkers that may contribute to increase diagnostic accuracy and help tailor therapeutic strategies in patients with neurodegenerative diseases.
Collapse
Affiliation(s)
- Luca Soraci
- Unit of Geriatric Medicine, Italian National Research Center of Aging (IRCCS INRCA), Cosenza, Italy.
| | - Andrea Corsonello
- Unit of Geriatric Medicine, Italian National Research Center of Aging (IRCCS INRCA), Cosenza, Italy.
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy.
| | - Ersilia Paparazzo
- Department of Biology, Ecology and Earth Sciences, University of Calabria, Rende, Italy.
| | - Alberto Montesanto
- Department of Biology, Ecology and Earth Sciences, University of Calabria, Rende, Italy.
| | - Francesco Piacenza
- Advanced Technology Center for Aging Research, Italian National Research Center of Aging (IRCCS INRCA), IRCCS INRCA, Ancona, Italy.
| | - Fabiola Olivieri
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Ancona, Italy.
- Clinic of Laboratory and Precision Medicine, Italian National Research Center of Aging (IRCCS INRCA), Ancona, Italy.
| | | | | | - Silvia Marino
- IRCCS Centro Neurolesi "Bonino-Pulejo”, Messina, Italy.
| | | | - Leonardo Biscetti
- Section of Neurology, Italian National Research Center on Aging (IRCCS INRCA), Ancona, Italy.
| |
Collapse
|
5
|
Leichtle A, Lupatsii M, Graspeuntner S, Jeschke S, Penxová Z, Kurabi A, Ryan AF, Rupp J, Pries R, Bruchhage KL. Anti-inflammatory response to 1,8-Cineol and associated microbial communities in Otitis media patients. Sci Rep 2024; 14:16362. [PMID: 39014066 PMCID: PMC11252366 DOI: 10.1038/s41598-024-67498-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 07/11/2024] [Indexed: 07/18/2024] Open
Abstract
Chronic Otitis Media (COM) is defined as long term inflammation and colonization with pathogenic bacteria due to a defect or retraction of the tympanic membrane. Surgical interventions are often augmented by antibiotic resistance development and therefore, off-label treatment using the natural drug 1,8-Cineol was carried out. All COM patients underwent antibiotic therapy and middle ear surgery and developed antibiotic resistances. Microbiological investigations from the auditory canal and stool samples were performed in correlation with the clinical course. Therapy of COM patients with 1,8-Cineol revealed a clear reduction of inflammatory microbes P. aeruginosa and Proteus mirabilis in ear samples as well as intestinal Prevotella copri, which was associated with an improved clinical outcome in certain individuals. The present off-label study revealed manifold anti-inflammatory effects of the natural monoterpene 1,8-Cineol in Otitis media patients. A better understanding of the underlying mechanisms will improve the current treatment options and possible forms of application of this natural drug.
Collapse
Affiliation(s)
- Anke Leichtle
- Department of Otorhinolaryngology, University of Luebeck, 23538, Lübeck, Germany
| | - Mariia Lupatsii
- Department of Infectious Diseases and Microbiology, University of Luebeck, Lübeck, Germany
| | - Simon Graspeuntner
- Department of Infectious Diseases and Microbiology, University of Luebeck, Lübeck, Germany
| | - Stephanie Jeschke
- Department of Otorhinolaryngology, University of Luebeck, 23538, Lübeck, Germany
| | - Zuzana Penxová
- Department of Otorhinolaryngology, University of Luebeck, 23538, Lübeck, Germany
| | - Arwa Kurabi
- Department of Surgery/ Otolaryngology, University of California San Diego, La Jolla, USA
| | - Allen Frederic Ryan
- Department of Surgery/ Otolaryngology, University of California San Diego, La Jolla, USA
| | - Jan Rupp
- Department of Infectious Diseases and Microbiology, University of Luebeck, Lübeck, Germany
| | - Ralph Pries
- Department of Otorhinolaryngology, University of Luebeck, 23538, Lübeck, Germany
| | | |
Collapse
|
6
|
Mahbub NU, Islam MM, Hong ST, Chung HJ. Dysbiosis of the gut microbiota and its effect on α-synuclein and prion protein misfolding: consequences for neurodegeneration. Front Cell Infect Microbiol 2024; 14:1348279. [PMID: 38435303 PMCID: PMC10904658 DOI: 10.3389/fcimb.2024.1348279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Accepted: 01/24/2024] [Indexed: 03/05/2024] Open
Abstract
Abnormal behavior of α-synuclein and prion proteins is the hallmark of Parkinson's disease (PD) and prion illnesses, respectively, being complex neurological disorders. A primary cause of protein aggregation, brain injury, and cognitive loss in prion illnesses is the misfolding of normal cellular prion proteins (PrPC) into an infectious form (PrPSc). Aggregation of α-synuclein causes disruptions in cellular processes in Parkinson's disease (PD), leading to loss of dopamine-producing neurons and motor symptoms. Alteration in the composition or activity of gut microbes may weaken the intestinal barrier and make it possible for prions to go from the gut to the brain. The gut-brain axis is linked to neuroinflammation; the metabolites produced by the gut microbiota affect the aggregation of α-synuclein, regulate inflammation and immunological responses, and may influence the course of the disease and neurotoxicity of proteins, even if their primary targets are distinct proteins. This thorough analysis explores the complex interactions that exist between the gut microbiota and neurodegenerative illnesses, particularly Parkinson's disease (PD) and prion disorders. The involvement of the gut microbiota, a complex collection of bacteria, archaea, fungi, viruses etc., in various neurological illnesses is becoming increasingly recognized. The gut microbiome influences neuroinflammation, neurotransmitter synthesis, mitochondrial function, and intestinal barrier integrity through the gut-brain axis, which contributes to the development and progression of disease. The review delves into the molecular mechanisms that underlie these relationships, emphasizing the effects of microbial metabolites such as bacterial lipopolysaccharides (LPS), and short-chain fatty acids (SCFAs) in regulating brain functioning. Additionally, it looks at how environmental influences and dietary decisions affect the gut microbiome and whether they could be risk factors for neurodegenerative illnesses. This study concludes by highlighting the critical role that the gut microbiota plays in the development of Parkinson's disease (PD) and prion disease. It also provides a promising direction for future research and possible treatment approaches. People afflicted by these difficult ailments may find hope in new preventive and therapeutic approaches if the role of the gut microbiota in these diseases is better understood.
Collapse
Affiliation(s)
- Nasir Uddin Mahbub
- Department of Biomedical Sciences and Institute for Medical Science, Jeonbuk National University Medical School, Jeonju, Republic of Korea
| | - Md Minarul Islam
- Department of Biomedical Sciences and Institute for Medical Science, Jeonbuk National University Medical School, Jeonju, Republic of Korea
| | - Seong-Tshool Hong
- Department of Biomedical Sciences and Institute for Medical Science, Jeonbuk National University Medical School, Jeonju, Republic of Korea
| | - Hea-Jong Chung
- Gwangju Center, Korea Basic Science Institute, Gwangju, Republic of Korea
| |
Collapse
|
7
|
Kiran NS, Yashaswini C, Chatterjee A. Noxious ramifications of cosmetic pollutants on gastrointestinal microbiome: A pathway to neurological disorders. Life Sci 2024; 336:122311. [PMID: 38043908 DOI: 10.1016/j.lfs.2023.122311] [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: 08/04/2023] [Revised: 11/10/2023] [Accepted: 11/24/2023] [Indexed: 12/05/2023]
Abstract
On exposure to cosmetic pollutants, gastrointestinal dysbiosis, which is characterised by a disturbance in the gut microbiota, has come into focus as a possible contributor to the occurrence of neurotoxic consequences. It is normal practice to use personal care products that include parabens, phthalates, sulphates, triclosans/triclocarbans and micro/nano plastics. These substances have been found in a variety of bodily fluids and tissues, demonstrating their systemic dispersion. Being exposed to these cosmetic pollutants has been linked in recent research to neurotoxicity, including cognitive decline and neurodevelopmental problems. A vital part of sustaining gut health and general well-being is the gut flora. Increased intestinal permeability, persistent inflammation, and impaired metabolism may result from disruption of the gut microbial environment, which may in turn contribute to neurotoxicity. The link between gastrointestinal dysbiosis and the neurotoxic effects brought on by cosmetic pollutants may be explained by a number of processes, primarily the gut-brain axis. For the purpose of creating preventative and therapeutic measures, it is crucial to comprehend the intricate interactions involving cosmetic pollutants, gastrointestinal dysbiosis, and neurotoxicity. This review provides an in-depth understanding of the various hazardous cosmetic pollutants and its potential role in the occurrence of neurological disorders via gastrointestinal dysbiosis, providing insights into various described and hypothetical mechanisms regarding the complex toxic effects of these industrial pollutants.
Collapse
Affiliation(s)
- Neelakanta Sarvashiva Kiran
- Department of Biotechnology, School of Applied Sciences, REVA University, Kattigenahalli, Yelahanka, Bangalore, Karnataka 560064, India
| | - Chandrashekar Yashaswini
- Department of Biotechnology, School of Applied Sciences, REVA University, Kattigenahalli, Yelahanka, Bangalore, Karnataka 560064, India
| | - Ankita Chatterjee
- Department of Biotechnology, School of Applied Sciences, REVA University, Kattigenahalli, Yelahanka, Bangalore, Karnataka 560064, India.
| |
Collapse
|
8
|
Su Y, Wang D, Liu N, Yang J, Sun R, Zhang Z. Clostridium butyricum improves cognitive dysfunction in ICV-STZ-induced Alzheimer's disease mice via suppressing TLR4 signaling pathway through the gut-brain axis. PLoS One 2023; 18:e0286086. [PMID: 37267300 DOI: 10.1371/journal.pone.0286086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Accepted: 05/08/2023] [Indexed: 06/04/2023] Open
Abstract
In recent years, the relationship between gut-brain axis and Alzheimer's disease (AD) attracted increasing attention. The aim of this study is to investigate the therapeutic effect of Clostridium butyricum (CB) on intraventricular injection of streptozotocin (ICV-STZ)-induced mice and the potential mechanisms. ICV-STZ mice were treated with CB by gavage for 21 consecutive days. The pharmacological effect of CB was assessed by behavior test, brain tissue H&E staining and tau protein phosphorylation levels of hippocampus tissues. The expression levels of TLR4, MYD88, NF-κB p65, TNF-α, iNOS, Occludin and ZO-1 in hippocampal and colonic tissues were detected by Western-blot method. 16S rRNA gene sequencing analysis was used to analyze the intestinal microbiota of mice. The results showed that CB improved the cognitive dysfunction of ICV-STZ mice, restored the structure and cell number of hippocampal and cortical neurons, decreased the protein levels of pSer404-tau protein in hippocampal tissues and TLR4, MYD88, NF-κB p65 and iNOS in hippocampal and colonic tissues, and increased the protein levels of Occludin and ZO-1 in colonic tissues. Meanwhile, CB reversed the changes of intestinal microbiota in AD mice. Therefore, the mechanisms of cognitive function and brain pathological changes in AD mice improved by CB may be related to the regulation of TLR4 signaling pathway and intestinal microbiota. This study supports the potential anti-AD effect of CB and initially revealed its pharmacological mechanism of CB, providing a theoretical basis for further clinical application of CB.
Collapse
Affiliation(s)
- Yunfang Su
- Henan Engineering Research Center for Prevention and Treatment of Major Chronic Diseases with Chinese Medicine, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan Province, China
- The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan Province, China
| | - Dahui Wang
- LiangShan College (LiShui) China, Lishui University, Lishui, Zhejiang Province, China
- Henan JinBaiHe Biotechnology Co. LTD, Nanjing, Jiangsu, China
| | - Ningning Liu
- Henan Engineering Research Center for Prevention and Treatment of Major Chronic Diseases with Chinese Medicine, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan Province, China
| | - Jiajia Yang
- School of Acupuncture and Massage, Henan University of Chinese Medicine, Zhengzhou, Henan Province, China
| | - Ruiqin Sun
- Henan Engineering Research Center for Prevention and Treatment of Major Chronic Diseases with Chinese Medicine, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan Province, China
| | - Zhenqiang Zhang
- Henan Engineering Research Center for Prevention and Treatment of Major Chronic Diseases with Chinese Medicine, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan Province, China
| |
Collapse
|
9
|
MicroRNAs and MAPKs: Evidence of These Molecular Interactions in Alzheimer's Disease. Int J Mol Sci 2023; 24:ijms24054736. [PMID: 36902178 PMCID: PMC10003111 DOI: 10.3390/ijms24054736] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 02/24/2023] [Accepted: 02/27/2023] [Indexed: 03/06/2023] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder known to be the leading cause of dementia worldwide. Many microRNAs (miRNAs) were found deregulated in the brain or blood of AD patients, suggesting a possible key role in different stages of neurodegeneration. In particular, mitogen-activated protein kinases (MAPK) signaling can be impaired by miRNA dysregulation during AD. Indeed, the aberrant MAPK pathway may facilitate the development of amyloid-beta (Aβ) and Tau pathology, oxidative stress, neuroinflammation, and brain cell death. The aim of this review was to describe the molecular interactions between miRNAs and MAPKs during AD pathogenesis by selecting evidence from experimental AD models. Publications ranging from 2010 to 2023 were considered, based on PubMed and Web of Science databases. According to obtained data, several miRNA deregulations may regulate MAPK signaling in different stages of AD and conversely. Moreover, overexpressing or silencing miRNAs involved in MAPK regulation was seen to improve cognitive deficits in AD animal models. In particular, miR-132 is of particular interest due to its neuroprotective functions by inhibiting Aβ and Tau depositions, as well as oxidative stress, through ERK/MAPK1 signaling modulation. However, further investigations are required to confirm and implement these promising results.
Collapse
|
10
|
Su Y, Liu N, Sun R, Ma J, Li Z, Wang P, Ma H, Sun Y, Song J, Zhang Z. Radix Rehmanniae Praeparata (Shu Dihuang) exerts neuroprotective effects on ICV-STZ-induced Alzheimer's disease mice through modulation of INSR/IRS-1/AKT/GSK-3β signaling pathway and intestinal microbiota. Front Pharmacol 2023; 14:1115387. [PMID: 36843923 PMCID: PMC9945319 DOI: 10.3389/fphar.2023.1115387] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Accepted: 01/27/2023] [Indexed: 02/10/2023] Open
Abstract
Radix Rehmanniae Praeparata (RRP, Shu Dihuang in Cinese) is widely used as primal medicine in Chinese herbal formula for the treatment of Alzheimer's disease (AD). However, the underlying mechanism of RRP for AD remains unclear. The aim of this study was to investigate the therapeutic effect of RRP on intracerebroventricular injection of streptozotocin (ICV-STZ)-induced AD model mice and its potential mechanism. ICV-STZ mice were continuously gavaged with RRP for 21 days. The pharmacological effects of RRP were evaluated by behavioral tests, brain tissue H&E staining and hippocampal tau protein phosphorylation levels. The expression levels of insulin receptor (INSR), IRS-1, pSer473-AKT/AKT and pSer9-GSK-3β/GSK-3β proteins in hippocampal and cortical tissues were detected by Western-blot method. The 16S rRNA gene sequencing was used to analyze the changes of intestinal microbiota in mice. The compounds in RRP were analyzed by mass spectrometry and their binding ability to INSR proteins was detected by molecular docking. The results showed that RRP ameliorated cognitive dysfunction and neuronal pathological changes of brain tissue in ICV-STZ mice, reduced tau protein hyperphosphorylation, INSR, IRS-1, pSer473-AKT/AKT, and pSer9-GSK-3β/GSK-3β levels in hippocampal and cortical tissues. Meanwhile, RRP reversed ICV-STZ-induced dysregulation of intestinal microbiota in AD mice. Mass spectrometry analysis showed that the RRP consisted mainly of seven compounds, namely Acteoside (Verbascoside), 5-Hydroxymethyl-2-furaldehyde (5-HMF), Apigenin7-O-glucuronide, Icariin, Gallic acid, Quercetin-3β-D-glucoside, and Geniposide. Molecular docking results further indicated that the compounds in RRP have binding ability to INSR protein and potential multiple synergistic effects. RRP ameliorates cognitive dysfunction and brain histopathological changes in AD mice. The mechanism of RRP ameliorating AD may be related to the regulation of INSR/IRS-1/AKT/GSK-3β signaling pathway and intestinal microbiota. This study supports the potential anti-AD efficacy of RRP and initially reveals the pharmacological mechanism of RRP, providing a theoretical basis for further clinical application of RRP.
Collapse
Affiliation(s)
- Yunfang Su
- Henan Engineering Research Center for Prevention and Treatment of Major Chronic Diseases with Chinese Medicine, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, China,The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Ningning Liu
- Henan Engineering Research Center for Prevention and Treatment of Major Chronic Diseases with Chinese Medicine, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, China
| | - Ruiqin Sun
- Henan Engineering Research Center for Prevention and Treatment of Major Chronic Diseases with Chinese Medicine, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, China
| | - Jinlian Ma
- Henan Engineering Research Center for Prevention and Treatment of Major Chronic Diseases with Chinese Medicine, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, China
| | - Zhonghua Li
- Henan Engineering Research Center for Prevention and Treatment of Major Chronic Diseases with Chinese Medicine, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, China
| | - Pan Wang
- Henan Engineering Research Center for Prevention and Treatment of Major Chronic Diseases with Chinese Medicine, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, China
| | - Huifen Ma
- Henan Engineering Research Center for Prevention and Treatment of Major Chronic Diseases with Chinese Medicine, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, China
| | - Yiran Sun
- Henan Engineering Research Center for Prevention and Treatment of Major Chronic Diseases with Chinese Medicine, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, China
| | - Junying Song
- Henan Engineering Research Center for Prevention and Treatment of Major Chronic Diseases with Chinese Medicine, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, China
| | - Zhenqiang Zhang
- Henan Engineering Research Center for Prevention and Treatment of Major Chronic Diseases with Chinese Medicine, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, China,*Correspondence: Zhenqiang Zhang,
| |
Collapse
|
11
|
L K, Ng TKS, Wee HN, Ching J. Gut-brain axis through the lens of gut microbiota and their relationships with Alzheimer's disease pathology: Review and recommendations. Mech Ageing Dev 2023; 211:111787. [PMID: 36736919 DOI: 10.1016/j.mad.2023.111787] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Revised: 01/05/2023] [Accepted: 01/30/2023] [Indexed: 02/04/2023]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder that affects millions of people worldwide. Growing evidence suggests that the gut microbiome (GM) plays a pivotal role in the pathogenesis of AD through the microbiota-gut-brain axis (MGB). Alterations in GM composition and diversity have been observed in both animal models and in human patients with AD. GM dysbiosis has been implicated in increased intestinal permeability, blood-brain barrier (BBB) impairment, neuroinflammation and the development of hallmarks of AD. Further elucidation of the role of GM in AD could pave way for the development of holistic predictive methods for determining AD risk and progression of disease. Furthermore, accumulating evidence suggests that GM modulation could alleviate adverse symptoms of AD or serve as a preventive measure. In addition, increasing evidence shows that Type 2 Diabetes Mellitus (T2DM) is often comorbid with AD, with common GM alterations and inflammatory response, which could chart the development of GM-related treatment interventions for both diseases. We conclude by exploring the therapeutic potential of GM in alleviating symptoms of AD and in reducing risk. Furthermore, we also propose future directions in AD research, namely fecal microbiota transplantation (FMT) and precision medicine.
Collapse
Affiliation(s)
- Krishaa L
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore
| | - Ted Kheng Siang Ng
- Arizona State University, Edson College of Nursing and Health Innovation, USA.
| | - Hai Ning Wee
- Cardiovascular and Metabolic Disorders Programme, Duke-NUS Medical School, Singapore
| | - Jianhong Ching
- Cardiovascular and Metabolic Disorders Programme, Duke-NUS Medical School, Singapore; KK Research Centre, KK Women's and Children's Hospital, Singapore.
| |
Collapse
|
12
|
Xu QQ, Su ZR, Yang W, Zhong M, Xian YF, Lin ZX. Patchouli alcohol attenuates the cognitive deficits in a transgenic mouse model of Alzheimer's disease via modulating neuropathology and gut microbiota through suppressing C/EBPβ/AEP pathway. J Neuroinflammation 2023; 20:19. [PMID: 36717922 PMCID: PMC9887791 DOI: 10.1186/s12974-023-02704-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 01/22/2023] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is a chronic neurodegenerative disease characterized by progressive cognitive dysfunctions and behavioral impairments. Patchouli alcohol (PA), isolated from Pogostemonis Herba, exhibits multiple pharmacological properties, including neuroprotective effects. This study aimed to investigate the therapeutic effects of PA against AD using the TgCRND8 transgenic AD mouse model, and to explore the underlying mechanisms targeting CCAAT/enhancer-binding protein β/asparagine endopeptidase (C/EBPβ/AEP) signaling pathway. METHODS After genotyping to confirm the transgenicity, drug treatments were administered intragastrically once daily to 3-month-old TgCRND8 mice for 4 consecutive months. Several behavioral tests were applied to assess different aspects of neurological functions. Then the brain and colon tissues were harvested for in-depth mechanistic studies. To further verify whether PA exerts anti-AD effects via modulating C/EBPβ/AEP signaling pathway in TgCRND8 mice, adeno-associated virus (AAV) vectors encoding CEBP/β were bilaterally injected into the hippocampal CA1 region in TgCRND8 mice to overexpress C/EBPβ. Additionally, the fecal microbiota transplantation (FMT) experiment was performed to verify the potential role of gut microbiota on the anti-AD effects of PA. RESULTS Our results showed that PA treatment significantly improved activities of daily living (ADL), ameliorated the anxiety-related behavioral deficits and cognitive impairments in TgCRND8 mice. PA modulated the amyloid precursor protein (APP) processing. PA also markedly reduced the levels of beta-amyloid (Aβ) 40 and Aβ42, suppressed Aβ plaque burdens, inhibited tau protein hyperphosphorylation at several sites and relieved neuroinflammation in the brains of TgCRND8 mice. Moreover, PA restored gut dysbiosis and inhibited the activation of the C/EBPβ/AEP signaling pathway in the brain and colon tissues of TgCRND8 mice. Interestingly, PA strikingly alleviated the AD-like pathologies induced by the overexpression of C/EBPβ in TgCRND8 mice. Additionally, the FMT of fecal microbiota from the PA-treated TgCRND8 mice significantly alleviated the cognitive impairments and AD-like pathologies in the germ-free TgCRND8 mice. CONCLUSION All these findings amply demonstrated that PA could ameliorate the cognitive deficits in TgCRND8 mice via suppressing Aβ plaques deposition, hyperphosphorylation of tau protein, neuroinflammation and gut dysbiosis through inhibiting the activation of C/EBPβ/AEP pathway, suggesting that PA is a promising naturally occurring chemical worthy of further development into the pharmaceutical treatment of AD.
Collapse
Affiliation(s)
- Qing-Qing Xu
- grid.10784.3a0000 0004 1937 0482School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, People’s Republic of China
| | - Zi-Ren Su
- grid.411866.c0000 0000 8848 7685Guangdong Provincial Key Laboratory of New Drug Development and Research of Chinese Medicine, Mathematical Engineering Academy of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006 China
| | - Wen Yang
- grid.10784.3a0000 0004 1937 0482School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, People’s Republic of China
| | - Mei Zhong
- grid.10784.3a0000 0004 1937 0482School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, People’s Republic of China
| | - Yan-Fang Xian
- grid.10784.3a0000 0004 1937 0482School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, People’s Republic of China
| | - Zhi-Xiu Lin
- grid.10784.3a0000 0004 1937 0482School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, People’s Republic of China ,grid.10784.3a0000 0004 1937 0482Hong Kong Institute of Integrative Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, People’s Republic of China ,grid.10784.3a0000 0004 1937 0482Li Dak Sum Yip Yio Chin R&D Centre for Chinese Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, People’s Republic of China
| |
Collapse
|
13
|
Bruchhage KL, Lupatsii M, Möllenkolk F, Leffers D, Kurabi A, Jürgens T, Graspeuntner S, Hollfelder D, Leichtle A. Hearing rehabilitation and microbial shift after middle ear surgery with Vibrant Soundbridge in patients with chronic otitis media. Eur Arch Otorhinolaryngol 2023; 280:3107-3118. [PMID: 36662266 DOI: 10.1007/s00405-022-07795-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 12/08/2022] [Indexed: 01/21/2023]
Abstract
INTRODUCTION Patients with otitis media (OM) encounter significant functional hearing impairment with conductive, or a combined hearing loss and long-term sequelae involving impaired speech/language development in children, reduced academic achievement and irreversible disorders of middle and inner ear requiring a long time therapy and/or multiple surgeries. In its persistent chronic form, Otitis media (COM) can often only be treated by undergoing ear surgery for hearing restoration. The persistent inflammatory reaction plays a major role, often caused by multi-resistant pathogens in the ear. Herein, we present outcomes of patients implanted with currently the only FDA approved active Middle Ear Implant Vibrant Soundbridge (VSB), suffering from persistent COM. METHODS The study enrolled 42 patients, treated by performing middle ear (ME) surgery to different extents and implanted with the VSB to various structures in the ME. Included were 17 children and 25 adults that had recurrent and/or persisting OM and significant hearing loss. Preoperative and postoperative patients' audiometric data were evaluated and the benefit with VSB assessed using the Glasgow Benefit Inventory for adults and pediatric cohorts. The microbial spectrum of pathogens was assessed before and after surgery, exploring the colonization of the otopathogens, as well as the intestinal microbiome from individually burdened patients. RESULTS The mean functional gain is 29.7 dB HL (range from 10 to 56.2 dB HL) with a significant improvement in speech intelligibility in quiet. Following VSB implantation, no significant differences in coupling were observed at low complication rates. Postoperatively patients showed significantly increased benefit with VSB compared to the untreated situation, including less otorrhea, pain, medical visits, and medication intake, with no recurrent OM and significant bacterial shift in otopathogens. The analysis of the intestinal microbiome displayed a high abundance of bacterial strains that might be linked to chronic and persistent inflammation. CONCLUSIONS Functional ear surgery including rehabilitation with a VSB in patients suffering from COM present to be safe and effective. The successful acceptance accompanied by the improved audiological performance resulted in significant benefit with VSB, with a shift in the ear pathogens and altered microbiome and thus is a great opportunity to be treated.
Collapse
Affiliation(s)
- Karl-Ludwig Bruchhage
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Schleswig-Holstein, Lübeck, Germany
| | - Mariia Lupatsii
- Department of Infectious Diseases and Microbiology, University Hospital Schleswig-Holstein, Lübeck, Germany
| | - Friederike Möllenkolk
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Schleswig-Holstein, Lübeck, Germany
| | - David Leffers
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Schleswig-Holstein, Lübeck, Germany
| | - Arwa Kurabi
- Division of Otolaryngology, Department of Surgery, UCSD School of Medicine, La Jolla, San Diego, USA
| | - Tim Jürgens
- Institute of Acoustics, University of Applied Sciences Lübeck, Lübeck, Germany
| | - Simon Graspeuntner
- Department of Infectious Diseases and Microbiology, University Hospital Schleswig-Holstein, Lübeck, Germany.,German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
| | - Daniela Hollfelder
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Schleswig-Holstein, Lübeck, Germany
| | - Anke Leichtle
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Schleswig-Holstein, Lübeck, Germany.
| |
Collapse
|
14
|
Kim JE, Roh YJ, Choi YJ, Lee SJ, Jin YJ, Song HJ, Seol AY, Son HJ, Hong JT, Hwang DY. Dysbiosis of Fecal Microbiota in Tg2576 Mice for Alzheimer's Disease during Pathological Constipation. Int J Mol Sci 2022; 23:ijms232314928. [PMID: 36499254 PMCID: PMC9736912 DOI: 10.3390/ijms232314928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 11/24/2022] [Accepted: 11/25/2022] [Indexed: 12/05/2022] Open
Abstract
Tg2576 transgenic mice for Alzheimer's disease (AD) exhibited significant phenotypes for neuropathological constipation, but no research has been conducted on the association of the fecal microbiota with dysbiosis. The correlation between fecal microbiota composition and neuropathological constipation in Tg2576 mice was investigated by examining the profile of fecal microbiota and fecal microbiota transplantation (FMT) in 9-10-month-old Tg2576 mice with the AD phenotypes and constipation. Several constipation phenotypes, including stool parameters, colon length, and histopathological structures, were observed prominently in Tg2576 mice compared to the wild-type (WT) mice. The fecal microbiota of Tg2576 mice showed decreases in Bacteroidetes and increases in the Firmicutes and Proteobacteria populations at the phylum level. The FMT study showed that stool parameters, including weight, water content, and morphology, decreased remarkably in the FMT group transplanted with a fecal suspension of Tg2576 mice (TgFMT) compared to the FMT group transplanted with a fecal suspension of WT mice (WFMT). The distribution of myenteric neurons and the interstitial cells of Cajal (ICC), as well as the enteric nervous system (ENS) function, remained lower in the TgFMT group. These results suggest that the neuropathological constipation phenotypes of Tg2576 mice may be tightly linked to the dysbiosis of the fecal microbiota.
Collapse
Affiliation(s)
- Ji-Eun Kim
- Department of Biomaterials Science (BK21 FOUR Program), Life and Industry Convergence Research Institute, College of Natural Resources and Life Science, Pusan National University, Miryang 50463, Republic of Korea
| | - Yu-Jeong Roh
- Department of Biomaterials Science (BK21 FOUR Program), Life and Industry Convergence Research Institute, College of Natural Resources and Life Science, Pusan National University, Miryang 50463, Republic of Korea
| | - Yun-Ju Choi
- Department of Biomaterials Science (BK21 FOUR Program), Life and Industry Convergence Research Institute, College of Natural Resources and Life Science, Pusan National University, Miryang 50463, Republic of Korea
| | - Su-Jin Lee
- Department of Biomaterials Science (BK21 FOUR Program), Life and Industry Convergence Research Institute, College of Natural Resources and Life Science, Pusan National University, Miryang 50463, Republic of Korea
| | - You-Jeong Jin
- Department of Biomaterials Science (BK21 FOUR Program), Life and Industry Convergence Research Institute, College of Natural Resources and Life Science, Pusan National University, Miryang 50463, Republic of Korea
| | - Hee-Jin Song
- Department of Biomaterials Science (BK21 FOUR Program), Life and Industry Convergence Research Institute, College of Natural Resources and Life Science, Pusan National University, Miryang 50463, Republic of Korea
| | - A-Yun Seol
- Department of Biomaterials Science (BK21 FOUR Program), Life and Industry Convergence Research Institute, College of Natural Resources and Life Science, Pusan National University, Miryang 50463, Republic of Korea
| | - Hong-Joo Son
- Department of Life Science and Environmental Biochemistry, Life and Industry Convergence Research Institute, College of Natural Resources and Life Science, Pusan National University, Miryang 50463, Republic of Korea
| | - Jin-Tae Hong
- College of Pharmacy, Chungbuk National University, Chungju 28644, Republic of Korea
| | - Dae-Youn Hwang
- Department of Biomaterials Science (BK21 FOUR Program), Life and Industry Convergence Research Institute, College of Natural Resources and Life Science, Pusan National University, Miryang 50463, Republic of Korea
- Longevity & Wellbeing Research Center, Laboratory Animals Resources Center, Pusan National University, Miryang 50463, Republic of Korea
- Correspondence: ; Tel.: +82-55-350-5388
| |
Collapse
|
15
|
Liu J, Tang W, Hou L, Wang J, Wang R, Zhang Y, Dong Z, Liu R, Yu S. Alteration of gut microbiota in migraine patients with irritable bowel syndrome in a Chinese Han population. Front Neurol 2022; 13:899056. [PMID: 36468070 PMCID: PMC9709108 DOI: 10.3389/fneur.2022.899056] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 10/26/2022] [Indexed: 09/10/2024] Open
Abstract
Objective Migraine is frequently reported in patients with irritable bowel syndrome (IBS), and emerging evidence suggests that gut microbiota plays a role in migraine and IBS. However, alterations in the gut microbiome in migraine patients with IBS remain unknown. This study aimed to explore the compositions of gut microbiota in migraine patients with IBS in a Chinese Han population. Methods Sixteen migraine patients with IBS and thirteen age- and gender-matched IBS patients with similar dietary and lifestyle habits were enrolled in this pilot study. Demographic data, clinical data, eating habits, lifestyle habits, comorbidities, and medications were recorded using a unified case registration form. Questionnaires for the Migraine Disability Assessment (MIDAS), Pittsburgh Sleep Quality Index (PSQI), Hamilton Anxiety Scale (HAMA), and Hamilton Depression Scale (HAMD) were completed. Fecal samples were collected, and microbial DNA was extracted. Gut microbiota 16S ribosomal RNA (16S rRNA) gene sequencing targeting the V4 region was performed using the Illumina HiSeq 2500 high-throughput sequencing platform. The relationships between gut microbiota and clinical characteristics of migraine were analyzed. Results The structure of gut microbiota differed between migraine patients with IBS and patients with IBS, while the richness and diversity of gut microbiota in migraine patients with IBS showed no significant difference from that of patients with IBS. We found a higher relative abundance of the genus Parabacteroides and a lower relative abundance of the genera Paraprevotella, Lachnospiraceae_UCG-010, Lactococcus, Collinsella, and Comamonas in migraine patients with IBS than in patients with IBS. According to random forest predictive models, the phylum Bacteroidota shows the most important role in migraine patients with IBS. Furthermore, no statistical correlation was found between significantly different taxa at the genus level and migraine clinical data. Conclusion This study identified that altered gut microbiota occurred in Chinese Han migraine patients with IBS, but no correlation was found between gut microbiota and the clinical characteristics of migraine. Further study is needed to better understand the role of gut microbiota in the pathogenesis of migraine in IBS.
Collapse
Affiliation(s)
- Jieqiong Liu
- Department of Neurology, The First Medical Center, Chinese PLA General Hospital, Chinese PLA Medical School, Beijing, China
- Department of Neurology, Cangzhou Central Hospital, Hebei Medical University, Cangzhou, China
| | - Wenjing Tang
- Department of Neurology, The First Medical Center, Chinese PLA General Hospital, Chinese PLA Medical School, Beijing, China
| | - Lei Hou
- Department of Neurology, The First Medical Center, Chinese PLA General Hospital, Chinese PLA Medical School, Beijing, China
| | - Jing Wang
- Department of Neurology, The First Medical Center, Chinese PLA General Hospital, Chinese PLA Medical School, Beijing, China
| | - Rongfei Wang
- Department of Neurology, The First Medical Center, Chinese PLA General Hospital, Chinese PLA Medical School, Beijing, China
| | - Yaofen Zhang
- Department of Neurology, The First Medical Center, Chinese PLA General Hospital, Chinese PLA Medical School, Beijing, China
| | - Zhao Dong
- Department of Neurology, The First Medical Center, Chinese PLA General Hospital, Chinese PLA Medical School, Beijing, China
| | - Ruozhuo Liu
- Department of Neurology, The First Medical Center, Chinese PLA General Hospital, Chinese PLA Medical School, Beijing, China
| | - Shengyuan Yu
- Department of Neurology, The First Medical Center, Chinese PLA General Hospital, Chinese PLA Medical School, Beijing, China
| |
Collapse
|
16
|
Oliveira MET, Paulino GVB, Dos Santos Júnior ED, da Silva Oliveira FA, Melo VMM, Ursulino JS, de Aquino TM, Shetty AK, Landell MF, Gitaí DLG. Multi-omic Analysis of the Gut Microbiome in Rats with Lithium-Pilocarpine-Induced Temporal Lobe Epilepsy. Mol Neurobiol 2022; 59:6429-6446. [PMID: 35962889 DOI: 10.1007/s12035-022-02984-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Accepted: 07/29/2022] [Indexed: 11/25/2022]
Abstract
Evidence supports that the gut microbiota and bacteria-dependent metabolites influence the maintenance of epileptic brain activity. However, the alterations in the gut microbiota between epileptic versus healthy individuals are poorly understood. We used a multi-omic approach to evaluate the changes in the composition of gut metagenome as well in the fecal metabolomic profile in rats before and after being submitted to status epilepticus (SE)-induced temporal lobe epilepsy (TLE). The 16S ribosomal RNA (rRNA) sequencing of fecal samples coupled to bioinformatic analysis revealed taxonomic, compositional, and functional shifts in epileptic rats. The species richness (Chao1 index) was significantly lower in the post-TLE group, and the β-diversity analysis revealed clustering separated from the pre-TLE group. The taxonomic abundance analysis showed a significant increase of phylum Desulfobacterota and a decrease of Patescibacteria in the post-TLE group. The DESEq2 and LEfSe analysis resulted in 18 genera significantly enriched between post-TLE and pre-TLE groups at the genus level. We observed that epileptic rats present a peculiar metabolic phenotype, including a lower concentration of D-glucose and L-lactic acid and a higher concentration of L-glutamic acid and glycine. The microbiota-host metabolic correlation analysis showed that the genera differentially abundant in post-TLE rats are associated with the altered metabolites, especially the proinflammatory Desulfovibrio and Marvinbryantia, which were enriched in epileptic animals and positively correlated with these excitatory neurotransmitters and carbohydrate metabolites. Therefore, our data revealed a correlation between dysbacteriosis in epileptic animals and fecal metabolites that are known to be relevant for maintaining epileptic brain activity by enhancing chronic inflammation, an excitatory-inhibitory imbalance, and/or a metabolic disturbance. These data are promising and suggest that targeting the gut microbiota could provide a novel avenue for preventing and treating acquired epilepsy. However, the causal relationship between these microbial/metabolite components and the SRS occurrence still needs further exploration.
Collapse
Affiliation(s)
- Maria Eduarda T Oliveira
- Laboratory of Cellular and Molecular Biology (LBCM), Institute of Biological and Health Sciences, Federal University of Alagoas, Maceió, AL, 57072-900, Brazil
| | - Gustavo V B Paulino
- Laboratory of Molecular Diversity (LDM), Institute of Biological and Health Sciences, Federal University of Alagoas, Maceió, AL, 57072-900, Brazil
| | - Erivaldo D Dos Santos Júnior
- Laboratory of Cellular and Molecular Biology (LBCM), Institute of Biological and Health Sciences, Federal University of Alagoas, Maceió, AL, 57072-900, Brazil
| | - Francisca A da Silva Oliveira
- Laboratory of Microbial Ecology and Biotechnology (Lembiotech), Department of Biology, Universidade Federal Do Ceará, Campus do Pici, Bloco 909, Fortaleza, CE, 60455-760, Brazil
| | - Vânia M M Melo
- Laboratory of Microbial Ecology and Biotechnology (Lembiotech), Department of Biology, Universidade Federal Do Ceará, Campus do Pici, Bloco 909, Fortaleza, CE, 60455-760, Brazil
| | - Jeferson S Ursulino
- Nucleus of Analysis and Research in Nuclear Magnetic Resonance - NAPRMN, Institute of Chemistry and Biotechnology, Federal University of Alagoas, Maceió, AL, 57072-900, Brazil
| | - Thiago M de Aquino
- Nucleus of Analysis and Research in Nuclear Magnetic Resonance - NAPRMN, Institute of Chemistry and Biotechnology, Federal University of Alagoas, Maceió, AL, 57072-900, Brazil
| | - Ashok K Shetty
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center College of Medicine, College Station, TX, USA
| | - Melissa Fontes Landell
- Laboratory of Molecular Diversity (LDM), Institute of Biological and Health Sciences, Federal University of Alagoas, Maceió, AL, 57072-900, Brazil.
| | - Daniel Leite Góes Gitaí
- Laboratory of Cellular and Molecular Biology (LBCM), Institute of Biological and Health Sciences, Federal University of Alagoas, Maceió, AL, 57072-900, Brazil.
| |
Collapse
|
17
|
Thu Thuy Nguyen V, Endres K. Targeting gut microbiota to alleviate neuroinflammation in Alzheimer's disease. Adv Drug Deliv Rev 2022; 188:114418. [PMID: 35787390 DOI: 10.1016/j.addr.2022.114418] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 06/27/2022] [Accepted: 06/28/2022] [Indexed: 02/08/2023]
Abstract
The gut microbiota came into focus within the last years regarding being associated with or even underlying neuropsychiatric diseases. The existence of the gut-brain-axis makes it highly plausible that bacterial metabolites or toxins that escape the intestinal environment or approach the vagal connections towards the brain, exert devastating effects on the central nervous system. In Alzheimer's disease (AD), growing evidence for dysbiotic changes in the gut microbiota is obtained, even though the question for cause or consequence remains open. Nevertheless, using modulation of microbiota to address inflammatory processes seems an attractive therapeutic approach as certain microbial products such as short chain fatty acids have been proven to exert beneficial cognitive effects. In this review, we summarize, contemporary knowledge on neuroinflammation and inflammatory processes within the brain and even more detailed in the gut in AD, try to conclude whom to target regarding human microbial commensals and report on current interventional trials.
Collapse
Affiliation(s)
- Vu Thu Thuy Nguyen
- Department of Psychiatry and Psychotherapy, University Medical Center Mainz, Johannes Gutenberg-University Mainz, Germany
| | - Kristina Endres
- Department of Psychiatry and Psychotherapy, University Medical Center Mainz, Johannes Gutenberg-University Mainz, Germany.
| |
Collapse
|
18
|
Patients with Infections of The Central Nervous System Have Lowered Gut Microbiota Alpha Diversity. Curr Issues Mol Biol 2022; 44:2903-2914. [PMID: 35877424 PMCID: PMC9318043 DOI: 10.3390/cimb44070200] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 06/27/2022] [Accepted: 06/27/2022] [Indexed: 11/17/2022] Open
Abstract
There are multiple lines of evidence for the existence of communication between the central nervous system (CNS), gut, and intestinal microbiome. Despite extensive analysis conducted on various neurological disorders, the gut microbiome was not yet analyzed in neuroinfections. In the current study, we analyzed the gut microbiome in 47 consecutive patients hospitalized with neuroinfection (26 patients had viral encephalitis/meningitis; 8 patients had bacterial meningitis) and in 20 matched for age and gender health controls. Using the QIIME pipeline, 16S rRNA sequencing and classification into operational taxonomic units (OTUs) were performed on the earliest stool sample available. Bacterial taxa such as Clostridium, Anaerostipes, Lachnobacterium, Lachnospira, and Roseburia were decreased in patients with neuroinfection when compared to controls. Alpha diversity metrics showed lower within-sample diversity in patients with neuroinfections, though there were no differences in beta diversity. Furthermore, there was no significant change by short-term (1-3 days) antibiotic treatment on the gut microbiota, although alpha diversity metrics, such as Chao1 and Shannon's index, were close to being statistically significant. The cause of differences between patients with neuroinfections and controls is unclear and could be due to inflammation accompanying the disease; however, the effect of diet modification and/or hospitalization cannot be excluded.
Collapse
|
19
|
Sittipo P, Choi J, Lee S, Lee YK. The function of gut microbiota in immune-related neurological disorders: a review. J Neuroinflammation 2022; 19:154. [PMID: 35706008 PMCID: PMC9199126 DOI: 10.1186/s12974-022-02510-1] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 06/01/2022] [Indexed: 12/13/2022] Open
Abstract
This review provides an overview of the importance of microbiota in the regulation of gut–brain communication in immune-related neurological disorders. The gastrointestinal (GI) tract hosts a diverse abundance of microbiota, referred to as gut microbiota. The gut microbiota plays a role in the maintenance of GI tract homeostasis and is likely to have multiple effects on brain development and function. The bidirectional communication between the gut microbiota and the brain is termed the microbiota–gut–brain axis. This communication between the intestine and the brain appears to affect human health and behavior, as certain animal studies have demonstrated the association between alterations in the gut microbiota and neurological disorders. Most insights about the microbiota–gut–brain axis come from germ-free animal models, which reveal the importance of gut microbiota in neural function. To date, many studies have observed the impact of the gut microbiota in patients with neurological disorders. Although many studies have investigated the microbiota–gut–brain axis, there are still limitations in translating this research to humans given the complexities of the relationship between the gut microbiota and the brain. In this review, we discuss emerging evidence of how the microbiota–gut–brain axis regulates brain development and function through biological networks, as well as the possible contribution of the microbiota–gut–brain axis in immune-related neurological disorders.
Collapse
Affiliation(s)
- Panida Sittipo
- Department of Integrated Biomedical Science, Soonchunhyang Institute of Medi-Bio Science, Soonchunhyang University, Cheonan, 31151, Republic of Korea
| | - Jaeyoon Choi
- Department of Integrated Biomedical Science, Soonchunhyang Institute of Medi-Bio Science, Soonchunhyang University, Cheonan, 31151, Republic of Korea
| | - Soojin Lee
- Department of Microbiology and Molecular Biology, Chungnam National University, Daejeon, 34134, Republic of Korea.
| | - Yun Kyung Lee
- Department of Integrated Biomedical Science, Soonchunhyang Institute of Medi-Bio Science, Soonchunhyang University, Cheonan, 31151, Republic of Korea.
| |
Collapse
|
20
|
Mou Y, Du Y, Zhou L, Yue J, Hu X, Liu Y, Chen S, Lin X, Zhang G, Xiao H, Dong B. Gut Microbiota Interact With the Brain Through Systemic Chronic Inflammation: Implications on Neuroinflammation, Neurodegeneration, and Aging. Front Immunol 2022; 13:796288. [PMID: 35464431 PMCID: PMC9021448 DOI: 10.3389/fimmu.2022.796288] [Citation(s) in RCA: 146] [Impact Index Per Article: 48.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Accepted: 02/22/2022] [Indexed: 02/05/2023] Open
Abstract
It has been noticed in recent years that the unfavorable effects of the gut microbiota could exhaust host vigor and life, yet knowledge and theory are just beginning to be established. Increasing documentation suggests that the microbiota-gut-brain axis not only impacts brain cognition and psychiatric symptoms but also precipitates neurodegenerative diseases, such as Alzheimer's disease (AD), Parkinson's disease (PD), and multiple sclerosis (MS). How the blood-brain barrier (BBB), a machinery protecting the central nervous system (CNS) from the systemic circulation, allows the risky factors derived from the gut to be translocated into the brain seems paradoxical. For the unique anatomical, histological, and immunological properties underpinning its permeable dynamics, the BBB has been regarded as a biomarker associated with neural pathogenesis. The BBB permeability of mice and rats caused by GM dysbiosis raises the question of how the GM and its metabolites change BBB permeability and causes the brain pathophysiology of neuroinflammation and neurodegeneration (NF&ND) and brain aging, a pivotal multidisciplinary field tightly associated with immune and chronic systemic inflammation. If not all, gut microbiota-induced systemic chronic inflammation (GM-SCI) mainly refers to excessive gut inflammation caused by gut mucosal immunity dysregulation, which is often influenced by dietary components and age, is produced at the interface of the intestinal barrier (IB) or exacerbated after IB disruption, initiates various common chronic diseases along its dispersal routes, and eventually impairs BBB integrity to cause NF&ND and brain aging. To illustrate the immune roles of the BBB in pathophysiology affected by inflammatory or "leaky" IB resulting from GM and their metabolites, we reviewed the selected publications, including the role of the BBB as the immune barrier, systemic chronic inflammation and inflammation influences on BBB permeability, NF&ND, and brain aging. To add depth to the bridging role of systemic chronic inflammation, a plausible mechanism indispensable for BBB corruption was highlighted; namely, BBB maintenance cues are affected by inflammatory cytokines, which may help to understand how GM and its metabolites play a major role in NF&ND and aging.
Collapse
Affiliation(s)
- Yi Mou
- Geroscience and Chronic Disease Department, The Eighth Municipal Hospital for the People, Chengdu, China
| | - Yu Du
- Department of Emergency and Critical Care Medicine, The Fourth West China Hospital, Sichuan University, Chengdu, China
| | - Lixing Zhou
- National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Jirong Yue
- National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Xianliang Hu
- Geroscience and Chronic Disease Department, The Eighth Municipal Hospital for the People, Chengdu, China
| | - Yixin Liu
- National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Sao Chen
- Geroscience and Chronic Disease Department, The Eighth Municipal Hospital for the People, Chengdu, China
| | - Xiufang Lin
- National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Gongchang Zhang
- National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Hengyi Xiao
- National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Birong Dong
- National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
21
|
Verhaar BJH, Hendriksen HMA, de Leeuw FA, Doorduijn AS, van Leeuwenstijn M, Teunissen CE, Barkhof F, Scheltens P, Kraaij R, van Duijn CM, Nieuwdorp M, Muller M, van der Flier WM. Gut Microbiota Composition Is Related to AD Pathology. Front Immunol 2022; 12:794519. [PMID: 35173707 PMCID: PMC8843078 DOI: 10.3389/fimmu.2021.794519] [Citation(s) in RCA: 78] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 12/31/2021] [Indexed: 12/26/2022] Open
Abstract
Introduction Several studies have reported alterations in gut microbiota composition of Alzheimer's disease (AD) patients. However, the observed differences are not consistent across studies. We aimed to investigate associations between gut microbiota composition and AD biomarkers using machine learning models in patients with AD dementia, mild cognitive impairment (MCI) and subjective cognitive decline (SCD). Materials and Methods We included 170 patients from the Amsterdam Dementia Cohort, comprising 33 with AD dementia (66 ± 8 years, 46%F, mini-mental state examination (MMSE) 21[19-24]), 21 with MCI (64 ± 8 years, 43%F, MMSE 27[25-29]) and 116 with SCD (62 ± 8 years, 44%F, MMSE 29[28-30]). Fecal samples were collected and gut microbiome composition was determined using 16S rRNA sequencing. Biomarkers of AD included cerebrospinal fluid (CSF) amyloid-beta 1-42 (amyloid) and phosphorylated tau (p-tau), and MRI visual scores (medial temporal atrophy, global cortical atrophy, white matter hyperintensities). Associations between gut microbiota composition and dichotomized AD biomarkers were assessed with machine learning classification models. The two models with the highest area under the curve (AUC) were selected for logistic regression, to assess associations between the 20 best predicting microbes and the outcome measures from these machine learning models while adjusting for age, sex, BMI, diabetes, medication use, and MMSE. Results The machine learning prediction for amyloid and p-tau from microbiota composition performed best with AUCs of 0.64 and 0.63. Highest ranked microbes included several short chain fatty acid (SCFA)-producing species. Higher abundance of [Clostridium] leptum and lower abundance of [Eubacterium] ventriosum group spp., Lachnospiraceae spp., Marvinbryantia spp., Monoglobus spp., [Ruminococcus] torques group spp., Roseburia hominis, and Christensenellaceae R-7 spp., was associated with higher odds of amyloid positivity. We found associations between lower abundance of Lachnospiraceae spp., Lachnoclostridium spp., Roseburia hominis and Bilophila wadsworthia and higher odds of positive p-tau status. Conclusions Gut microbiota composition was associated with amyloid and p-tau status. We extend on recent studies that observed associations between SCFA levels and AD CSF biomarkers by showing that lower abundances of SCFA-producing microbes were associated with higher odds of positive amyloid and p-tau status.
Collapse
Affiliation(s)
- Barbara J. H. Verhaar
- Department of Internal Medicine - Geriatrics, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center (UMC), Amsterdam, Netherlands
- Department of Internal and Vascular Medicine, Amsterdam University Medical Center (UMC), Amsterdam, Netherlands
- Alzheimer Center, Department of Neurology, Amsterdam Neuroscience, Amsterdam University Medical Center (UMC), Amsterdam, Netherlands
| | - Heleen M. A. Hendriksen
- Alzheimer Center, Department of Neurology, Amsterdam Neuroscience, Amsterdam University Medical Center (UMC), Amsterdam, Netherlands
| | - Francisca A. de Leeuw
- Alzheimer Center, Department of Neurology, Amsterdam Neuroscience, Amsterdam University Medical Center (UMC), Amsterdam, Netherlands
| | - Astrid S. Doorduijn
- Alzheimer Center, Department of Neurology, Amsterdam Neuroscience, Amsterdam University Medical Center (UMC), Amsterdam, Netherlands
| | - Mardou van Leeuwenstijn
- Alzheimer Center, Department of Neurology, Amsterdam Neuroscience, Amsterdam University Medical Center (UMC), Amsterdam, Netherlands
| | - Charlotte E. Teunissen
- Department of Clinical Chemistry, Amsterdam University Medical Center (UMC), Amsterdam, Netherlands
| | - Frederik Barkhof
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Center (UMC), Amsterdam, Netherlands
- University College London (UCL) Institutes of Neurology, Faculty of Brain Sciences, London, United Kingdom
| | - Philip Scheltens
- Alzheimer Center, Department of Neurology, Amsterdam Neuroscience, Amsterdam University Medical Center (UMC), Amsterdam, Netherlands
| | - Robert Kraaij
- Department of Internal Medicine, Erasmus Medical Center (MC), Rotterdam, Netherlands
| | - Cornelia M. van Duijn
- Department of Epidemiology, Erasmus Medical Center (MC), Rotterdam, Netherlands
- Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Max Nieuwdorp
- Department of Internal and Vascular Medicine, Amsterdam University Medical Center (UMC), Amsterdam, Netherlands
| | - Majon Muller
- Department of Internal Medicine - Geriatrics, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center (UMC), Amsterdam, Netherlands
| | - Wiesje M. van der Flier
- Alzheimer Center, Department of Neurology, Amsterdam Neuroscience, Amsterdam University Medical Center (UMC), Amsterdam, Netherlands
- Department of Epidemiology and Data Science, Amsterdam University Medical Center (UMC), Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
22
|
Bou Zerdan M, Hebbo E, Hijazi A, El Gemayel M, Nasr J, Nasr D, Yaghi M, Bouferraa Y, Nagarajan A. The Gut Microbiome and Alzheimer's Disease: A Growing Relationship. Curr Alzheimer Res 2022; 19:808-818. [PMID: 36578263 DOI: 10.2174/1567205020666221227090125] [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: 07/26/2022] [Revised: 10/03/2022] [Accepted: 10/19/2022] [Indexed: 12/30/2022]
Abstract
Evidence that the gut microbiota plays a key role in the pathogenesis of Alzheimer's disease is already unravelling. The microbiota-gut-brain axis is a bidirectional communication system that is not fully understood but includes neural, immune, endocrine, and metabolic pathways. The progression of Alzheimer's disease is supported by mechanisms related to the imbalance in the gut microbiota and the development of amyloid plaques in the brain, which are at the origin of Alzheimer's disease. Alterations in the composition of the gut microbiome led to dysregulation in the pathways governing this system. This leads to neurodegeneration through neuroinflammation and neurotransmitter dysregulation. Neurodegeneration and disruption of the blood-brain barrier are frontiers at the origin of Alzheimer's disease. Furthermore, bacteria populating the gut microbiota can secrete large amounts of amyloid proteins and lipopolysaccharides, which modulate signaling pathways and alter the production of proinflammatory cytokines associated with the pathogenesis of Alzheimer's disease. Importantly, through molecular mimicry, bacterial amyloids may elicit cross-seeding of misfolding and induce microglial priming at different levels of the brain-gut-microbiota axis. The potential mechanisms of amyloid spreading include neuron-to-neuron or distal neuron spreading, direct blood-brain barrier crossing, or via other cells such as astrocytes, fibroblasts, microglia, and immune system cells. Gut microbiota metabolites, including short-chain fatty acids, pro-inflammatory factors, and neurotransmitters may also affect AD pathogenesis and associated cognitive decline. The purpose of this review is to summarize and discuss the current findings that may elucidate the role of gut microbiota in the development of Alzheimer's disease. Understanding the underlying mechanisms may provide new insights into novel therapeutic strategies for Alzheimer's disease, such as probiotics and targeted oligosaccharides.
Collapse
Affiliation(s)
- Maroun Bou Zerdan
- Department of Internal Medicine, SUNY Upstate Medical University, New York, USA
- Department of Hematology and Oncology, Cleveland Clinic Florida, Weston, Florida, USA
| | - Elsa Hebbo
- Faculty of Medicine, American University of Beirut, Beirut 2020, Lebanon
| | - Ali Hijazi
- Faculty of Medicine, American University of Beirut, Beirut 2020, Lebanon
| | - Maria El Gemayel
- Department of Gastroenterology and Hepatology, Faculty of Medicine, Hotel-Dieu de France Hospital, Saint Joseph University, Beirut, Lebanon
| | - Janane Nasr
- Faculty of Medicine, Saint George Hospital, University of Balamand, Beirut, 1100, Lebanon
| | - Dayana Nasr
- Department of Internal Medicine, SUNY Upstate Medical University, New York, USA
| | - Marita Yaghi
- Department of Hematology and Oncology, Cleveland Clinic Florida, Weston, Florida, USA
| | - Youssef Bouferraa
- Department of Internal Medicine, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Arun Nagarajan
- Department of Hematology/Oncology, Cleveland Clinic, Weston, FL, 33331, USA
| |
Collapse
|
23
|
Wu D, Zhang Y, Dong S, Zhong C. Mutual interaction of microbiota and host immunity during health and diseases. BIOPHYSICS REPORTS 2021; 7:326-340. [PMID: 37287759 PMCID: PMC10233470 DOI: 10.52601/bpr.2021.200045] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 06/17/2021] [Indexed: 06/09/2023] Open
Abstract
Microbiota-host interaction has attracted more and more attentions in recent years. The association between microbiota and host health is largely attributed to its influence on host immune system. Microbial-derived antigens and metabolites play a critical role in shaping the host immune system, including regulating its development, activation, and function. However, during various diseases the microbiota-host communication is frequently found to be disordered. In particular, gut microbiota dysbiosis associated with or led to the occurrence and progression of infectious diseases, autoimmune diseases, metabolic diseases, and neurological diseases. Pathogenic microbes and their metabolites disturb the protective function of immune system, and lead to disordered immune responses that usually correlate with disease exacerbation. In the other hand, the immune system also regulates microbiota composition to keep host homeostasis. Here, we will discuss the current advances of our knowledge about the interactions between microbiota and host immune system during health and diseases.
Collapse
Affiliation(s)
- Di Wu
- Institute of Systems Biomedicine, Department of Immunology, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, Beijing 100191, China
| | - Yinlian Zhang
- Institute of Systems Biomedicine, Department of Immunology, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, Beijing 100191, China
| | - Suwei Dong
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Chemical Biology, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Chao Zhong
- Institute of Systems Biomedicine, Department of Immunology, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, Beijing 100191, China
| |
Collapse
|
24
|
Pogue AI, Lukiw WJ. microRNA-146a-5p, Neurotropic Viral Infection and Prion Disease (PrD). Int J Mol Sci 2021; 22:ijms22179198. [PMID: 34502105 PMCID: PMC8431499 DOI: 10.3390/ijms22179198] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 08/09/2021] [Accepted: 08/16/2021] [Indexed: 12/15/2022] Open
Abstract
The human brain and central nervous system (CNS) harbor a select sub-group of potentially pathogenic microRNAs (miRNAs), including a well-characterized NF-kB-sensitive Homo sapiens microRNA hsa-miRNA-146a-5p (miRNA-146a). miRNA-146a is significantly over-expressed in progressive and often lethal viral- and prion-mediated and related neurological syndromes associated with progressive inflammatory neurodegeneration. These include ~18 different viral-induced encephalopathies for which data are available, at least ~10 known prion diseases (PrD) of animals and humans, Alzheimer’s disease (AD) and other sporadic and progressive age-related neurological disorders. Despite the apparent lack of nucleic acids in prions, both DNA- and RNA-containing viruses along with prions significantly induce miRNA-146a in the infected host, but whether this represents part of the host’s adaptive immunity, innate-immune response or a mechanism to enable the invading prion or virus a successful infection is not well understood. Current findings suggest an early and highly interactive role for miRNA-146a: (i) as a major small noncoding RNA (sncRNA) regulator of innate-immune responses and inflammatory signaling in cells of the human brain and CNS; (ii) as a critical component of the complement system and immune-related neurological dysfunction; (iii) as an inducible sncRNA of the brain and CNS that lies at a critical intersection of several important neurobiological adaptive immune response processes with highly interactive associations involving complement factor H (CFH), Toll-like receptor pathways, the innate-immunity, cytokine production, apoptosis and neural cell decline; and (iv) as a potential biomarker for viral infection, TSE and AD and other neurological diseases in both animals and humans. In this report, we review the recent data supporting the idea that miRNA-146a may represent a novel and unique sncRNA-based biomarker for inflammatory neurodegeneration in multiple species. This paper further reviews the current state of knowledge regarding the nature and mechanism of miRNA-146a in viral and prion infection of the human brain and CNS with reference to AD wherever possible.
Collapse
Affiliation(s)
| | - Walter J. Lukiw
- LSU Neuroscience Center, Louisiana State University Health Science Center, New Orleans, LA 70112, USA
- Department of Ophthalmology, Louisiana State University Health Science Center, New Orleans, LA 70112, USA
- Department of Neurology, Louisiana State University Health Science Center, New Orleans, LA 70112, USA
- Correspondence:
| |
Collapse
|
25
|
Blood-Based Biomarkers of Neuroinflammation in Alzheimer's Disease: A Central Role for Periphery? Diagnostics (Basel) 2021; 11:diagnostics11091525. [PMID: 34573867 PMCID: PMC8464786 DOI: 10.3390/diagnostics11091525] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/17/2021] [Accepted: 08/20/2021] [Indexed: 12/12/2022] Open
Abstract
Neuroinflammation represents a central feature in the development of Alzheimer’s disease (AD). The resident innate immune cells of the brain are the principal players in neuroinflammation, and their activation leads to a defensive response aimed at promoting β-amyloid (Aβ) clearance. However, it is now widely accepted that the peripheral immune system—by virtue of a dysfunctional blood–brain barrier (BBB)—is involved in the pathogenesis and progression of AD; microglial and astrocytic activation leads to the release of chemokines able to recruit peripheral immune cells into the central nervous system (CNS); at the same time, cytokines released by peripheral cells are able to cross the BBB and act upon glial cells, modifying their phenotype. To successfully fight this neurodegenerative disorder, accurate and sensitive biomarkers are required to be used for implementing an early diagnosis, monitoring the disease progression and treatment effectiveness. Interestingly, as a result of the bidirectional communication between the brain and the periphery, the blood compartment ends up reflecting several pathological changes occurring in the AD brain and can represent an accessible source for such biomarkers. In this review, we provide an overview on some of the most promising peripheral biomarkers of neuroinflammation, discussing their pathogenic role in AD.
Collapse
|
26
|
Vitamin K2 Holds Promise for Alzheimer's Prevention and Treatment. Nutrients 2021; 13:nu13072206. [PMID: 34199021 PMCID: PMC8308377 DOI: 10.3390/nu13072206] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 06/24/2021] [Accepted: 06/24/2021] [Indexed: 12/24/2022] Open
Abstract
Recent studies have highlighted the importance of vitamin K2 (VK2) in human health. However, there have been no clinical studies investigating the role of VK2 in the prevention or treatment of Alzheimer's disease (AD), a debilitating disease for which currently there is no cure. In reviewing basic science research and clinical studies that have connected VK2 to factors involved in AD pathogenesis, we have found a growing body of evidence demonstrating that VK2 has the potential to slow the progression of AD and contribute to its prevention. In our review, we consider the antiapoptotic and antioxidant effects of VK2 and its impact on neuroinflammation, mitochondrial dysfunction, cognition, cardiovascular health, and comorbidities in AD. We also examine the link between dysbiosis and VK2 in the context of the microbiome's role in AD pathogenesis. Our review is the first to consider the physiological roles of VK2 in the context of AD, and, given the recent shift in AD research toward nonpharmacological interventions, our findings emphasize the timeliness and need for clinical studies involving VK2.
Collapse
|