1
|
Shen X, Mu X. Systematic Insights into the Relationship between the Microbiota-Gut-Brain Axis and Stroke with the Focus on Tryptophan Metabolism. Metabolites 2024; 14:399. [PMID: 39195495 DOI: 10.3390/metabo14080399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 07/15/2024] [Accepted: 07/15/2024] [Indexed: 08/29/2024] Open
Abstract
Stroke, as a serious cerebral vascular disease with high incidence and high rates of disability and mortality, has limited therapeutic options due to the narrow time window. Compelling evidence has highlighted the significance of the gut microbiota and gut-brain axis as critical regulatory factors affecting stroke. Along the microbiota-gut-brain axis, tryptophan metabolism further acquires increasing attention for its intimate association with central nervous system diseases. For the purpose of exploring the potential role of tryptophan metabolism in stroke and providing systematic insights into the intricate connection of the microbiota-gut-brain axis with the pathological procedure of stroke, this review first summarized the practical relationship between microbiota and stroke by compiling the latest case-control research. Then, the microbiota-gut-brain axis, as well as its interaction with stroke, were comprehensively elucidated on the basis of the basic anatomical structure and physiological function. Based on the crosstalk of microbiota-gut-brain, we further focused on the tryptophan metabolism from the three major metabolic pathways, namely, the kynurenine pathway, serotonin pathway, and microbial pathway, within the axis. Moreover, the effects of tryptophan metabolism on stroke were appreciated and elaborated here, which is scarcely found in other reviews. Hopefully, the systematic illustration of the mechanisms and pathways along the microbiota-gut-brain axis will inspire more translational research from metabolic perspectives, along with more attention paid to tryptophan metabolism as a promising pharmaceutical target in order to reduce the risk of stroke, mitigate the stroke progression, and ameliorate the stroke prognosis.
Collapse
Affiliation(s)
- Xinyu Shen
- Genomics Research Center, Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province, College of Pharmacy, Harbin Medical University, Harbin 150081, China
- Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin 150081, China
| | - Xiaoqin Mu
- Genomics Research Center, Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province, College of Pharmacy, Harbin Medical University, Harbin 150081, China
- Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin 150081, China
| |
Collapse
|
2
|
Han B, Zhou S, Zhang Y, Chen S, Xi W, Liu C, Zhou X, Yuan M, Yu X, Li L, Wang Y, Ren H, Xie J, Li B, Ju M, Zhou Y, Liu Z, Xiong Z, Shen L, Zhang Y, Bai Y, Chen J, Jiang W, Yao H. Integrating spatial and single-cell transcriptomics to characterize the molecular and cellular architecture of the ischemic mouse brain. Sci Transl Med 2024; 16:eadg1323. [PMID: 38324639 DOI: 10.1126/scitranslmed.adg1323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 01/12/2024] [Indexed: 02/09/2024]
Abstract
Neuroinflammation is acknowledged as a pivotal pathological event after cerebral ischemia. However, there is limited knowledge of the molecular and spatial characteristics of nonneuronal cells, as well as of the interactions between cell types in the ischemic brain. Here, we used spatial transcriptomics to study the ischemic hemisphere in mice after stroke and sequenced the transcriptomes of 19,777 spots, allowing us to both visualize the transcriptional landscape within the tissue and identify gene expression profiles linked to specific histologic entities. Cell types identified by single-cell RNA sequencing confirmed and enriched the spatial annotation of ischemia-associated gene expression in the peri-infarct area of the ischemic hemisphere. Analysis of ligand-receptor interactions in cell communication revealed galectin-9 to cell-surface glycoprotein CD44 (LGALS9-CD44) as a critical signaling pathway after ischemic injury and identified microglia and macrophages as the main source of galectins after stroke. Extracellular vesicle-mediated Lgals9 delivery improved the long-term functional recovery in photothrombotic stroke mice. Knockdown of Cd44 partially reversed these therapeutic effects, inhibiting oligodendrocyte differentiation and remyelination. In summary, our study provides a detailed molecular and cellular characterization of the peri-infact area in a murine stroke model and revealed Lgals9 as potential treatment target that warrants further investigation.
Collapse
Affiliation(s)
- Bing Han
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China
| | - Shunheng Zhou
- Department of Biomedical Engineering, College of Automation Engineering, Nanjing University of Aeronautics and Astronautics, Nanjing 211106, China
| | - Yuan Zhang
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China
| | - Sina Chen
- Department of Biomedical Engineering, College of Automation Engineering, Nanjing University of Aeronautics and Astronautics, Nanjing 211106, China
| | - Wen Xi
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China
| | - Chenchen Liu
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China
| | - Xu Zhou
- Department of Biomedical Engineering, College of Automation Engineering, Nanjing University of Aeronautics and Astronautics, Nanjing 211106, China
| | - Mengqin Yuan
- Department of Biomedical Engineering, College of Automation Engineering, Nanjing University of Aeronautics and Astronautics, Nanjing 211106, China
| | - Xiaoyu Yu
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China
| | - Lu Li
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China
| | - Yu Wang
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China
| | - Hui Ren
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China
| | - Jian Xie
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China
| | - Bin Li
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China
| | - Minzi Ju
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China
| | - You Zhou
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China
| | - Ziqi Liu
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China
| | - Zhongli Xiong
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China
| | - Ling Shen
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China
| | - Yuan Zhang
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China
| | - Ying Bai
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China
| | - Jun Chen
- Pittsburgh Institute of Brain Disorders and Recovery and Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA 15261, USA
| | - Wei Jiang
- Department of Biomedical Engineering, College of Automation Engineering, Nanjing University of Aeronautics and Astronautics, Nanjing 211106, China
| | - Honghong Yao
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China
- Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226019, China
- Institute of Life Sciences, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing 210009, China
| |
Collapse
|
3
|
Zhao L, Xiao J, Li S, Guo Y, Fu R, Hua S, Du Y, Xu S. The interaction between intestinal microenvironment and stroke. CNS Neurosci Ther 2023; 29 Suppl 1:185-199. [PMID: 37309254 PMCID: PMC10314114 DOI: 10.1111/cns.14275] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 05/09/2023] [Accepted: 05/10/2023] [Indexed: 06/14/2023] Open
Abstract
BACKGROUND Stroke is not only a major cause of disability but also the third leading cause of death, following heart disease and cancer. It has been established that stroke causes permanent disability in 80% of survivors. However, current treatment options for this patient population are limited. Inflammation and immune response are major features that are well-recognized to occur after a stroke. The gastrointestinal tract hosts complex microbial communities, the largest pool of immune cells, and forms a bidirectional regulation brain-gut axis with the brain. Recent experimental and clinical studies have highlighted the importance of the relationship between the intestinal microenvironment and stroke. Over the years, the influence of the intestine on stroke has emerged as an important and dynamic research direction in biology and medicine. AIMS In this review, we describe the structure and function of the intestinal microenvironment and highlight its cross-talk relationship with stroke. In addition, we discuss potential strategies aiming to target the intestinal microenvironment during stroke treatment. CONCLUSION The structure and function of the intestinal environment can influence neurological function and cerebral ischemic outcome. Improving the intestinal microenvironment by targeting the gut microbiota may be a new direction in treating stroke.
Collapse
Affiliation(s)
- Linna Zhao
- First Teaching Hospital of Tianjin University of Traditional Chinese MedicineTianjinChina
- National Clinical Research Center for Chinese Medicine Acupuncture and MoxibustionFirst Teaching Hospital of Tianjin University of Traditional Chinese MedicineTianjinChina
- Tianjin Key Laboratory of Translational Research of TCM Prescription and SyndromeTianjinChina
| | - Jie Xiao
- First Teaching Hospital of Tianjin University of Traditional Chinese MedicineTianjinChina
- National Clinical Research Center for Chinese Medicine Acupuncture and MoxibustionFirst Teaching Hospital of Tianjin University of Traditional Chinese MedicineTianjinChina
- Tianjin University of Traditional Chinese MedicineTianjinChina
| | - Songlin Li
- First Teaching Hospital of Tianjin University of Traditional Chinese MedicineTianjinChina
- National Clinical Research Center for Chinese Medicine Acupuncture and MoxibustionFirst Teaching Hospital of Tianjin University of Traditional Chinese MedicineTianjinChina
- Tianjin University of Traditional Chinese MedicineTianjinChina
| | - Yuying Guo
- First Teaching Hospital of Tianjin University of Traditional Chinese MedicineTianjinChina
- National Clinical Research Center for Chinese Medicine Acupuncture and MoxibustionFirst Teaching Hospital of Tianjin University of Traditional Chinese MedicineTianjinChina
- Tianjin Key Laboratory of Translational Research of TCM Prescription and SyndromeTianjinChina
| | - Rong Fu
- First Teaching Hospital of Tianjin University of Traditional Chinese MedicineTianjinChina
- National Clinical Research Center for Chinese Medicine Acupuncture and MoxibustionFirst Teaching Hospital of Tianjin University of Traditional Chinese MedicineTianjinChina
- Tianjin University of Traditional Chinese MedicineTianjinChina
| | - Shengyu Hua
- Tianjin University of Traditional Chinese MedicineTianjinChina
| | - Yuzheng Du
- First Teaching Hospital of Tianjin University of Traditional Chinese MedicineTianjinChina
- National Clinical Research Center for Chinese Medicine Acupuncture and MoxibustionFirst Teaching Hospital of Tianjin University of Traditional Chinese MedicineTianjinChina
| | - Shixin Xu
- First Teaching Hospital of Tianjin University of Traditional Chinese MedicineTianjinChina
- National Clinical Research Center for Chinese Medicine Acupuncture and MoxibustionFirst Teaching Hospital of Tianjin University of Traditional Chinese MedicineTianjinChina
- Tianjin Key Laboratory of Translational Research of TCM Prescription and SyndromeTianjinChina
| |
Collapse
|
4
|
Fernández-Calle R, Konings SC, Frontiñán-Rubio J, García-Revilla J, Camprubí-Ferrer L, Svensson M, Martinson I, Boza-Serrano A, Venero JL, Nielsen HM, Gouras GK, Deierborg T. APOE in the bullseye of neurodegenerative diseases: impact of the APOE genotype in Alzheimer's disease pathology and brain diseases. Mol Neurodegener 2022; 17:62. [PMID: 36153580 PMCID: PMC9509584 DOI: 10.1186/s13024-022-00566-4] [Citation(s) in RCA: 94] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 08/29/2022] [Indexed: 02/06/2023] Open
Abstract
ApoE is the major lipid and cholesterol carrier in the CNS. There are three major human polymorphisms, apoE2, apoE3, and apoE4, and the genetic expression of APOE4 is one of the most influential risk factors for the development of late-onset Alzheimer's disease (AD). Neuroinflammation has become the third hallmark of AD, together with Amyloid-β plaques and neurofibrillary tangles of hyperphosphorylated aggregated tau protein. This review aims to broadly and extensively describe the differential aspects concerning apoE. Starting from the evolution of apoE to how APOE's single-nucleotide polymorphisms affect its structure, function, and involvement during health and disease. This review reflects on how APOE's polymorphisms impact critical aspects of AD pathology, such as the neuroinflammatory response, particularly the effect of APOE on astrocytic and microglial function and microglial dynamics, synaptic function, amyloid-β load, tau pathology, autophagy, and cell-cell communication. We discuss influential factors affecting AD pathology combined with the APOE genotype, such as sex, age, diet, physical exercise, current therapies and clinical trials in the AD field. The impact of the APOE genotype in other neurodegenerative diseases characterized by overt inflammation, e.g., alpha- synucleinopathies and Parkinson's disease, traumatic brain injury, stroke, amyotrophic lateral sclerosis, and multiple sclerosis, is also addressed. Therefore, this review gathers the most relevant findings related to the APOE genotype up to date and its implications on AD and CNS pathologies to provide a deeper understanding of the knowledge in the APOE field.
Collapse
Affiliation(s)
- Rosalía Fernández-Calle
- Department of Experimental Medical Science, Experimental Neuroinflammation Laboratory, Lund University, Lund, Sweden
| | - Sabine C. Konings
- Department of Experimental Medical Science, Experimental Dementia Research Unit, Lund University, Lund, Sweden
| | - Javier Frontiñán-Rubio
- Oxidative Stress and Neurodegeneration Group, Faculty of Medicine, Universidad de Castilla-La Mancha, Ciudad Real, Spain
| | - Juan García-Revilla
- Department of Experimental Medical Science, Experimental Neuroinflammation Laboratory, Lund University, Lund, Sweden
- Departamento de Bioquímica Y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, and Instituto de Biomedicina de Sevilla-Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - Lluís Camprubí-Ferrer
- Department of Experimental Medical Science, Experimental Neuroinflammation Laboratory, Lund University, Lund, Sweden
| | - Martina Svensson
- Department of Experimental Medical Science, Experimental Neuroinflammation Laboratory, Lund University, Lund, Sweden
| | - Isak Martinson
- Department of Experimental Medical Science, Experimental Neuroinflammation Laboratory, Lund University, Lund, Sweden
| | - Antonio Boza-Serrano
- Department of Experimental Medical Science, Experimental Neuroinflammation Laboratory, Lund University, Lund, Sweden
- Departamento de Bioquímica Y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, and Instituto de Biomedicina de Sevilla-Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - José Luís Venero
- Departamento de Bioquímica Y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, and Instituto de Biomedicina de Sevilla-Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - Henrietta M. Nielsen
- Department of Biochemistry and Biophysics at, Stockholm University, Stockholm, Sweden
| | - Gunnar K. Gouras
- Department of Experimental Medical Science, Experimental Dementia Research Unit, Lund University, Lund, Sweden
| | - Tomas Deierborg
- Department of Experimental Medical Science, Experimental Neuroinflammation Laboratory, Lund University, Lund, Sweden
| |
Collapse
|
5
|
Ohlsson L, Isaxon C, Wrighton S, El Ouahidi W, Fornell L, Uller L, Ansar S, Voss U. Short-term exposure to urban PM 2.5 particles induces histopathological and inflammatory changes in the rat small intestine. Physiol Rep 2022; 10:e15249. [PMID: 35416410 PMCID: PMC9006536 DOI: 10.14814/phy2.15249] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 03/08/2022] [Accepted: 03/15/2022] [Indexed: 06/14/2023] Open
Abstract
Air pollution and exposure to fine airborne particles with aerodynamic diameter <2.5 μm (PM2.5 ) negatively impacts human health. Airways constitute a primary route of exposure but PM2.5 -contaminated food, drinks as well as mucociliary and hepatobiliary clearance all constitute potential entry points into the intestine. This study evaluated intestinal histopathological and inflammatory changes as well as enteric neuronal numbers after short- or long-term exposure to urban PM2.5 . Using a nebulizer, male rats were exposed to a mist with a concentration of 5.3mg PM2.5 /m3 for 8 h (short term) or 1.8 mg PM2.5 /m3 for 3 h/day, 5 days/week for 8 weeks (long-term) with controls run in parallel. Samples were taken from three regions of the small intestine as well as the colon. Results showed that short-term exposure to PM2.5 induces mucosal lesions and reduces IL1β levels in the small intestine but not colon. No significant changes were observed after long-term exposure, suggesting the presence of intestinal adaptation to environmental stressors in the PM2.5 . To our knowledge, this is the first study to systematically characterize regional effects along the intestine.
Collapse
Affiliation(s)
- Lena Ohlsson
- Unit of Experimental Vascular ResearchDepartment of Clinical SciencesLund UniversityLundSweden
| | - Christina Isaxon
- Division of Ergonomics and Aerosol TechnologyDepartment of Design SciencesLund UniversityLundSweden
| | - Sebastian Wrighton
- Division of Infection MedicineDepartment of Clinical SciencesLund UniversityLundSweden
| | - Wissal El Ouahidi
- Unit of Applied Neurovascular ResearchDepartment of Clinical SciencesLund UniversityLundSweden
| | - Lisa Fornell
- Unit of Applied Neurovascular ResearchDepartment of Clinical SciencesLund UniversityLundSweden
| | - Lena Uller
- Unit of Respiratory ImmunopharmacologyDepartment of Experimental Medical SciencesLund UniversityLundSweden
| | - Saema Ansar
- Unit of Applied Neurovascular ResearchDepartment of Clinical SciencesLund UniversityLundSweden
| | - Ulrikke Voss
- Unit of Applied Neurovascular ResearchDepartment of Clinical SciencesLund UniversityLundSweden
| |
Collapse
|
6
|
Irradiation Induces Tuft Cell Hyperplasia and Myenteric Neuronal Loss in the Absence of Dietary Fiber in a Mouse Model of Pelvic Radiotherapy. GASTROENTEROLOGY INSIGHTS 2022. [DOI: 10.3390/gastroent13010010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Pelvic radiotherapy is associated with chronic intestinal dysfunction. Dietary approaches, such as fiber enrichment during and after pelvic radiotherapy, have been suggested to prevent or reduce dysfunctions. In the present paper, we aimed to investigate whether a diet rich in fermentable fiber could have a positive effect on radiation-induced intestinal damage, especially focusing on tuft cells and enteric neurons. Male C57BL/6 mice were fed either a purified non-fiber diet or the same purified diet with 5% or 15% oat fiber added, starting two weeks prior to sham-irradiation or irradiation with four fractions of 8 Gray. The animals continued on the diets for 1, 6 or 18 weeks, after which the gross morphology of the colorectum was assessed together with the numbers of enteric neurons, tuft cells and crypt-surface units. The results showed that dietary fiber significantly affected the intestinal morphometrics, both in the short and long-term. The presence of dietary fiber stimulated the re-emergence of crypt-surface unit structures after irradiation. At 18 weeks, the animals fed with the non-fiber diet displayed more myenteric neurons than the animals fed with the dietary fibers, but irradiation resulted in a loss of neurons in the non-fiber fed animals. Irradiation, but not diet, affected the tuft cell numbers, and a significant increase in tuft cells was found 6 and 18 weeks after irradiation. In conclusion, dietary fiber intake has the potential to modify neuronal pathogenesis in the colorectum after irradiation. The long-lasting increase in tuft cells induced by irradiation may reflect an as yet unknown role in the mucosal pathophysiology after pelvic irradiation.
Collapse
|
7
|
Kramer P. Mitochondria-Microbiota Interaction in Neurodegeneration. Front Aging Neurosci 2022; 13:776936. [PMID: 35002678 PMCID: PMC8733591 DOI: 10.3389/fnagi.2021.776936] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 11/30/2021] [Indexed: 11/13/2022] Open
Abstract
Alzheimer’s and Parkinson’s are the two best-known neurodegenerative diseases. Each is associated with the excessive aggregation in the brain and elsewhere of its own characteristic amyloid proteins. Yet the two afflictions have much in common and often the same amyloids play a role in both. These amyloids need not be toxic and can help regulate bile secretion, synaptic plasticity, and immune defense. Moreover, when they do form toxic aggregates, amyloids typically harm not just patients but their pathogens too. A major port of entry for pathogens is the gut. Keeping the gut’s microbe community (microbiota) healthy and under control requires that our cells’ main energy producers (mitochondria) support the gut-blood barrier and immune system. As we age, these mitochondria eventually succumb to the corrosive byproducts they themselves release, our defenses break down, pathogens or their toxins break through, and the side effects of inflammation and amyloid aggregation become problematic. Although it gets most of the attention, local amyloid aggregation in the brain merely points to a bigger problem: the systemic breakdown of the entire human superorganism, exemplified by an interaction turning bad between mitochondria and microbiota.
Collapse
Affiliation(s)
- Peter Kramer
- Department of General Psychology, University of Padua, Padua, Italy
| |
Collapse
|
8
|
Abstract
The enteric nervous system (ENS) is the largest division of the peripheral nervous system and closely resembles components and functions of the central nervous system. Although the central role of the ENS in congenital enteric neuropathic disorders, including Hirschsprung disease and inflammatory and functional bowel diseases, is well acknowledged, its role in systemic diseases is less understood. Evidence of a disordered ENS has accumulated in neurodegenerative diseases ranging from amyotrophic lateral sclerosis, Alzheimer disease and multiple sclerosis to Parkinson disease as well as neurodevelopmental disorders such as autism. The ENS is a key modulator of gut barrier function and a regulator of enteric homeostasis. A 'leaky gut' represents the gateway for bacterial and toxin translocation that might initiate downstream processes. Data indicate that changes in the gut microbiome acting in concert with the individual genetic background can modify the ENS, central nervous system and the immune system, impair barrier function, and contribute to various disorders such as irritable bowel syndrome, inflammatory bowel disease or neurodegeneration. Here, we summarize the current knowledge on the role of the ENS in gastrointestinal and systemic diseases, highlighting its interaction with various key players involved in shaping the phenotypes. Finally, current flaws and pitfalls related to ENS research in addition to future perspectives are also addressed.
Collapse
|
9
|
do Carmo Neto JR, Braga YLL, da Costa AWF, Lucio FH, do Nascimento TC, dos Reis MA, Celes MRN, de Oliveira FA, Machado JR, da Silva MV. Biomarkers and Their Possible Functions in the Intestinal Microenvironment of Chagasic Megacolon: An Overview of the (Neuro)inflammatory Process. J Immunol Res 2021; 2021:6668739. [PMID: 33928170 PMCID: PMC8049798 DOI: 10.1155/2021/6668739] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Revised: 03/08/2021] [Accepted: 03/19/2021] [Indexed: 12/13/2022] Open
Abstract
The association between inflammatory processes and intestinal neuronal destruction during the progression of Chagasic megacolon is well established. However, many other components play essential roles, both in the long-term progression and control of the clinical status of patients infected with Trypanosoma cruzi. Components such as neuronal subpopulations, enteric glial cells, mast cells and their proteases, and homeostasis-related proteins from several organic systems (serotonin and galectins) are differentially involved in the progression of Chagasic megacolon. This review is aimed at revealing the characteristics of the intestinal microenvironment found in Chagasic megacolon by using different types of already used biomarkers. Information regarding these components may provide new therapeutic alternatives and improve the understanding of the association between T. cruzi infection and immune, endocrine, and neurological system changes.
Collapse
Affiliation(s)
- José Rodrigues do Carmo Neto
- Department of Bioscience and Technology, Institute of Tropical Pathology and Public Health, Federal University of Goiás, Goiânia, GO, Brazil
| | - Yarlla Loyane Lira Braga
- Department of Bioscience and Technology, Institute of Tropical Pathology and Public Health, Federal University of Goiás, Goiânia, GO, Brazil
| | - Arthur Wilson Florêncio da Costa
- Department of Bioscience and Technology, Institute of Tropical Pathology and Public Health, Federal University of Goiás, Goiânia, GO, Brazil
| | - Fernanda Hélia Lucio
- Department of Bioscience and Technology, Institute of Tropical Pathology and Public Health, Federal University of Goiás, Goiânia, GO, Brazil
| | - Thais Cardoso do Nascimento
- Department of Bioscience and Technology, Institute of Tropical Pathology and Public Health, Federal University of Goiás, Goiânia, GO, Brazil
| | - Marlene Antônia dos Reis
- Department of General Pathology, Federal University of Triângulo Mineiro, Uberaba, Minas Gerais, Brazil
| | - Mara Rubia Nunes Celes
- Department of Bioscience and Technology, Institute of Tropical Pathology and Public Health, Federal University of Goiás, Goiânia, GO, Brazil
| | - Flávia Aparecida de Oliveira
- Department of Bioscience and Technology, Institute of Tropical Pathology and Public Health, Federal University of Goiás, Goiânia, GO, Brazil
| | - Juliana Reis Machado
- Department of Bioscience and Technology, Institute of Tropical Pathology and Public Health, Federal University of Goiás, Goiânia, GO, Brazil
- Department of General Pathology, Federal University of Triângulo Mineiro, Uberaba, Minas Gerais, Brazil
| | - Marcos Vinícius da Silva
- Department of Microbiology, Immunology and Parasitology, Institute of Biological and Natural Sciences, Federal University of Triângulo Mineiro, Uberaba, Minas Gerais, Brazil
| |
Collapse
|
10
|
Wei M, Huang Q, Liu Z, Luo Y, Xia J. Intestinal Barrier Dysfunction Participates in the Pathophysiology of Ischemic Stroke. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2021; 20:401-416. [PMID: 33749565 DOI: 10.2174/1871527320666210322115808] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 10/12/2020] [Accepted: 11/30/2020] [Indexed: 11/22/2022]
Abstract
The gastrointestinal tract is a major organ for the body to absorb nutrients, water and electrolytes. At the same time, it is a tight barrier to resist the invasion of harmful substances and maintain the homeostasis of the internal environment. Destruction of the intestinal barrier is linked to the digestive system, cardiovascular system, endocrine system and other systemic diseases. Mounting evidence suggests that ischemic stroke not only changes the intestinal microbes, but also increases the permeability of the intestinal barrier, leading to bacterial translocation, infection, and even sepsis. The intestinal barrier, as part of the gut-brain axis, has also been proven to participate in the pathophysiological process of ischemic stroke. However, little attention has been paid to it. Since ischemic stroke is a major public health issue worldwide, there is an urgent need to know more about the disease for better prevention, treatment and prognosis. Therefore, understanding the pathophysiological relationship between ischemic stroke and the intestinal barrier will help researchers further uncover the pathophysiological mechanism of ischemic stroke and provide a novel therapeutic target for the treatment of ischemic stroke. Here, we review the physiology and pathology between ischemic stroke and intestinal barrier based on related articles published in the past ten years about the relationship between ischemic stroke, stroke risk factors and intestinal flora, intestinal barrier, and discuss the following parts: the intestinal barrier; possible mechanisms of intestinal barrier destruction in ischemic stroke; intestinal barrier destruction caused by stroke-related risk factors; intestinal barrier dysfunction in ischemic stroke; targeting the intestinal barrier to improve stroke; conclusions and perspectives.
Collapse
Affiliation(s)
- Minping Wei
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan 410008. China
| | - Qin Huang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan 410008. China
| | - Zeyu Liu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan 410008. China
| | - Yunfang Luo
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan 410008. China
| | - Jian Xia
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan 410008. China
| |
Collapse
|
11
|
Zhuang JJ, Zhou L, Zheng YH, Ding YS. The serum galectin-3 levels are associated with the severity and prognosis of ischemic stroke. Aging (Albany NY) 2021; 13:7454-7464. [PMID: 33686023 PMCID: PMC7993705 DOI: 10.18632/aging.202610] [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: 08/13/2020] [Accepted: 09/29/2020] [Indexed: 12/16/2022]
Abstract
Galectin-3, a microglia/macrophage-derived inflammatory mediator, plays a role in the stroke progression. In this single-center prospective study, we included 288 consecutive patients with a first-ever acute ischemic stroke to assess the association between galectin-3 serum level and clinical severity at admission and outcome at discharge by univariate and multivariate logistic regression. The results were presented as odds ratios (OR) and 95% confidence intervals (CI). Patients with high severity and poor outcomes had higher serum levels of galectin-3 (P<0.001 and P<0.001). Multivariate analysis suggested that a galectin-3 serum level in the highest quartile (The lowest three quartiles[Q1-3] as the reference) was associated with poor functional outcome (OR, 3.15; 95% CI, 2.44–3.87). The AUC (standard error) for the NIHSS and the combined model were 0.764 (0.031) and 0.823 (0.027), corresponding to a difference of 0.059 (0.004). This study shows that higher serum levels of galectin-3 are associated with stroke severity at admission and stroke prognosis at discharge in ischemic stroke.
Collapse
Affiliation(s)
- Jia-Jun Zhuang
- Department of Neurology, Weifang People's Hospital, Weifang, Shandong, China
| | - Li Zhou
- Department of Neurology, Weifang People's Hospital, Weifang, Shandong, China
| | - Yan-Hua Zheng
- Department of Neurology, Weifang People's Hospital, Weifang, Shandong, China
| | - Yan-Sheng Ding
- Department of Clinical Laboratory, Weifang People's Hospital, Weifang, Shandong, China
| |
Collapse
|
12
|
Mikaelyan KA, Krylov KY, Petrova MV, Shestopalov AE. [Intestine morphology and microbiocenosis changes in critically ill patients in neurosurgery]. ZHURNAL VOPROSY NEĬROKHIRURGII IMENI N. N. BURDENKO 2021; 85:104-110. [PMID: 33560626 DOI: 10.17116/neiro202185011104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
In recent years, the effect of critical conditions on intestine and the role of such changes in maintenance and progression of systemic disorders are of particular attention. This issue is relevant in critically ill neurosurgical patients too. Intestine morphology and microbiome changes in these patients represent a wide field for researches in intensive care and prevention of secondary damage to other organs and systems. This review ensures a current approach to the problem of intestine morphology and microbiome changes in critically ill neurosurgical patients. We reviewed the data from clinical studies and experiments reproducing a critical condition in animals. Most publications are indexed in the PubMed, e-library, Google Scholar databases. We also analyzed the data from NEJM, JAMA, Lancet, Critical Care and other issues. The manuscript contains an overview of 44 foreign and 13 domestic references; over 50% of researches were published within the past 5 years. Searching depth was over 50 years.
Collapse
Affiliation(s)
- K A Mikaelyan
- Russian Peoples' Friendship University, Moscow, Russia
| | - K Yu Krylov
- Pirogov Russian National Research Medical University, Moscow, Russia
| | - M V Petrova
- Russian Peoples' Friendship University, Moscow, Russia
| | - A E Shestopalov
- Federal Research Clinical Center of Intensive Care and Rehabilitation, Lytkino, Russia
| |
Collapse
|
13
|
Muralitharan RR, Jama HA, Xie L, Peh A, Snelson M, Marques FZ. Microbial Peer Pressure: The Role of the Gut Microbiota in Hypertension and Its Complications. HYPERTENSION (DALLAS, TEX. : 1979) 2020; 76:1674-1687. [PMID: 33012206 DOI: 10.1161/hypertensionaha.120.14473] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
There is increasing evidence of the influence of the gut microbiota on hypertension and its complications, such as chronic kidney disease, stroke, heart failure, and myocardial infarction. This is not surprising considering that the most common risk factors for hypertension, such as age, sex, medication, and diet, can also impact the gut microbiota. For example, sodium and fermentable fiber have been studied in relation to both hypertension and the gut microbiota. By combining second- and, now, third-generation sequencing with metabolomics approaches, metabolites, such as short-chain fatty acids and trimethylamine N-oxide, and their producers, have been identified and are now known to affect host physiology and the cardiovascular system. The receptors that bind these metabolites have also been explored with positive findings-examples include known short-chain fatty acid receptors, such as G-protein coupled receptors GPR41, GPR43, GPR109a, and OLF78 in mice. GPR41 and OLF78 have been shown to have inverse roles in blood pressure regulation, whereas GPR43 and GPR109A have to date been demonstrated to impact cardiac function. New treatment options in the form of prebiotics (eg, dietary fiber), probiotics (eg, Lactobacillus spp.), and postbiotics (eg, the short-chain fatty acids acetate, propionate, and butyrate) have all been demonstrated to be beneficial in lowering blood pressure in animal models, but the underlying mechanisms remain poorly understood and translation to hypertensive patients is still lacking. Here, we review the evidence for the role of the gut microbiota in hypertension, its risk factors, and cardiorenal complications and identify future directions for this exciting and fast-evolving field.
Collapse
Affiliation(s)
- Rikeish R Muralitharan
- From the Hypertension Research Laboratory, School of Biological Sciences, Faculty of Science (R.R.M., H.A.J., L.X., A.P., F.Z.M.), Monash University, Melbourne, Australia
- Institute for Medical Research, Ministry of Health Malaysia, Kuala Lumpur, Malaysia (R.R.M.)
| | - Hamdi A Jama
- From the Hypertension Research Laboratory, School of Biological Sciences, Faculty of Science (R.R.M., H.A.J., L.X., A.P., F.Z.M.), Monash University, Melbourne, Australia
- Heart Failure Research Group, Baker Heart and Diabetes Institute, Melbourne, Australia (H.A.J., F.Z.M.)
| | - Liang Xie
- From the Hypertension Research Laboratory, School of Biological Sciences, Faculty of Science (R.R.M., H.A.J., L.X., A.P., F.Z.M.), Monash University, Melbourne, Australia
- Department of Microbiology, Biomedicine Discovery Institute, Monash University, Clayton, Australia (L.X.)
| | - Alex Peh
- From the Hypertension Research Laboratory, School of Biological Sciences, Faculty of Science (R.R.M., H.A.J., L.X., A.P., F.Z.M.), Monash University, Melbourne, Australia
| | - Matthew Snelson
- Department of Diabetes, Central Clinical School (M.S.), Monash University, Melbourne, Australia
| | - Francine Z Marques
- From the Hypertension Research Laboratory, School of Biological Sciences, Faculty of Science (R.R.M., H.A.J., L.X., A.P., F.Z.M.), Monash University, Melbourne, Australia
- Heart Failure Research Group, Baker Heart and Diabetes Institute, Melbourne, Australia (H.A.J., F.Z.M.)
| |
Collapse
|
14
|
Li XJ, You XY, Wang CY, Li XL, Sheng YY, Zhuang PW, Zhang YJ. Bidirectional Brain-gut-microbiota Axis in increased intestinal permeability induced by central nervous system injury. CNS Neurosci Ther 2020; 26:783-790. [PMID: 32472633 PMCID: PMC7366750 DOI: 10.1111/cns.13401] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 03/19/2020] [Accepted: 04/28/2020] [Indexed: 12/14/2022] Open
Abstract
Central nervous system injuries may lead to the disorders of the hypothalamic‐pituitary‐adrenal axis, autonomic nervous system, and enteric nervous system. These effects then cause the changes in the intestinal microenvironment, such as a disordered intestinal immune system as well as alterations of intestinal bacteria. Ultimately, this leads to an increase in intestinal permeability. Inflammatory factors produced by the interactions between intestinal neurons and immune cells as well as the secretions and metabolites of intestinal flora can then migrate through the intestinal barrier, which will aggravate any peripheral inflammation and the central nervous system injury. The brain‐gut‐microbiota axis is a complex system that plays a crucial role in the occurrence and development of central nervous system diseases. It may also increase the consequences of preventative treatment. In this context, here we have summarized the factors that can lead to the increased intestinal permeability and some of the possible outcomes.
Collapse
Affiliation(s)
- Xiao-Jin Li
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xin-Yu You
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Cong-Ying Wang
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xue-Li Li
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yuan-Yuan Sheng
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Peng-Wei Zhuang
- Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin Key Laboratory of Chinese Medicine Pharmacology, Tianjin, China
| | - Yan-Jun Zhang
- Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin Key Laboratory of Chinese Medicine Pharmacology, Tianjin, China
| |
Collapse
|
15
|
Srejovic I, Selakovic D, Jovicic N, Jakovljević V, Lukic ML, Rosic G. Galectin-3: Roles in Neurodevelopment, Neuroinflammation, and Behavior. Biomolecules 2020; 10:biom10050798. [PMID: 32455781 PMCID: PMC7277476 DOI: 10.3390/biom10050798] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/1970] [Revised: 05/09/2020] [Accepted: 05/13/2020] [Indexed: 12/16/2022] Open
Abstract
There is a plethora of evidence to suggest that Galectin-3 plays an important role in normal functions of mammalian cells, as well as in different pathogenic conditions. This review highlights recent data published by researchers, including our own team, on roles of Galectin-3 in the nervous system. Here, we discuss the roles of Galectin-3 in brain development, its roles in glial cells, as well as the interactions of glial cells with other neural and invading cells in pathological conditions. Galectin-3 plays an important role in the pathogenesis of neuroinflammatory and neurodegenerative disorders, such as multiple sclerosis, Alzheimer’s disease, Parkinson’s disease, and Huntington’s disease. On the other hand, there is also evidence of the protective role of Galectin-3 due to its anti-apoptotic effect in target cells. Interestingly, genetic deletion of Galectin-3 affects behavioral patterns in maturing and adult mice. The results reviewed in this paper and recent development of highly specific inhibitors suggests that Galectin-3 may be an important therapeutic target in pathological conditions including the disorders of the central nervous system.
Collapse
Affiliation(s)
- Ivan Srejovic
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovica 69, 34000 Kragujevac Serbia; (I.S.); (D.S.); (V.J.)
| | - Dragica Selakovic
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovica 69, 34000 Kragujevac Serbia; (I.S.); (D.S.); (V.J.)
| | - Nemanja Jovicic
- Department of Histology and Embryology, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovica 69, 34000 Kragujevac, Serbia;
| | - Vladimir Jakovljević
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovica 69, 34000 Kragujevac Serbia; (I.S.); (D.S.); (V.J.)
- Department of Human Pathology, 1st Moscow State Medical University IM Sechenov, 119146 Moscow, Russia
| | - Miodrag L. Lukic
- Department of Physiology—Molecular Medicine Unit, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovica 69, 34000 Kragujevac, Serbia
- Correspondence: (M.L.L.); (G.R.)
| | - Gvozden Rosic
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovica 69, 34000 Kragujevac Serbia; (I.S.); (D.S.); (V.J.)
- Correspondence: (M.L.L.); (G.R.)
| |
Collapse
|
16
|
Cirillo C, Brihmat N, Castel-Lacanal E, Le Friec A, Barbieux-Guillot M, Raposo N, Pariente J, Viguier A, Simonetta-Moreau M, Albucher JF, Olivot JM, Desmoulin F, Marque P, Chollet F, Loubinoux I. Post-stroke remodeling processes in animal models and humans. J Cereb Blood Flow Metab 2020; 40:3-22. [PMID: 31645178 PMCID: PMC6928555 DOI: 10.1177/0271678x19882788] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 08/28/2019] [Accepted: 09/05/2019] [Indexed: 01/05/2023]
Abstract
After cerebral ischemia, events like neural plasticity and tissue reorganization intervene in lesioned and non-lesioned areas of the brain. These processes are tightly related to functional improvement and successful rehabilitation in patients. Plastic remodeling in the brain is associated with limited spontaneous functional recovery in patients. Improvement depends on the initial deficit, size, nature and localization of the infarction, together with the sex and age of the patient, all of them affecting the favorable outcome of reorganization and repair of damaged areas. A better understanding of cerebral plasticity is pivotal to design effective therapeutic strategies. Experimental models and clinical studies have fueled the current understanding of the cellular and molecular processes responsible for plastic remodeling. In this review, we describe the known mechanisms, in patients and animal models, underlying cerebral reorganization and contributing to functional recovery after ischemic stroke. We also discuss the manipulations and therapies that can stimulate neural plasticity. We finally explore a new topic in the field of ischemic stroke pathophysiology, namely the brain-gut axis.
Collapse
Affiliation(s)
- Carla Cirillo
- Toulouse NeuroImaging Center (ToNIC), INSERM, University Paul Sabatier, UPS, Toulouse, France
| | - Nabila Brihmat
- Toulouse NeuroImaging Center (ToNIC), INSERM, University Paul Sabatier, UPS, Toulouse, France
| | - Evelyne Castel-Lacanal
- Toulouse NeuroImaging Center (ToNIC), INSERM, University Paul Sabatier, UPS, Toulouse, France
| | - Alice Le Friec
- Toulouse NeuroImaging Center (ToNIC), INSERM, University Paul Sabatier, UPS, Toulouse, France
| | | | - Nicolas Raposo
- Toulouse NeuroImaging Center (ToNIC), INSERM, University Paul Sabatier, UPS, Toulouse, France
| | - Jérémie Pariente
- Toulouse NeuroImaging Center (ToNIC), INSERM, University Paul Sabatier, UPS, Toulouse, France
| | - Alain Viguier
- Toulouse NeuroImaging Center (ToNIC), INSERM, University Paul Sabatier, UPS, Toulouse, France
| | - Marion Simonetta-Moreau
- Toulouse NeuroImaging Center (ToNIC), INSERM, University Paul Sabatier, UPS, Toulouse, France
| | - Jean-François Albucher
- Toulouse NeuroImaging Center (ToNIC), INSERM, University Paul Sabatier, UPS, Toulouse, France
| | - Jean-Marc Olivot
- Toulouse NeuroImaging Center (ToNIC), INSERM, University Paul Sabatier, UPS, Toulouse, France
| | - Franck Desmoulin
- Toulouse NeuroImaging Center (ToNIC), INSERM, University Paul Sabatier, UPS, Toulouse, France
| | - Philippe Marque
- Toulouse NeuroImaging Center (ToNIC), INSERM, University Paul Sabatier, UPS, Toulouse, France
| | - François Chollet
- Toulouse NeuroImaging Center (ToNIC), INSERM, University Paul Sabatier, UPS, Toulouse, France
| | - Isabelle Loubinoux
- Toulouse NeuroImaging Center (ToNIC), INSERM, University Paul Sabatier, UPS, Toulouse, France
| |
Collapse
|
17
|
Yazar HO, Yazar T, Cihan M. A preliminary data: Evaluation of serum Galectin-3 levels in patients with Idiopathic Parkinson's Disease. J Clin Neurosci 2019; 70:164-168. [PMID: 31471077 DOI: 10.1016/j.jocn.2019.08.032] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 08/05/2019] [Indexed: 12/11/2022]
Abstract
AIM In our study, we aimed to collect data for the hypothesis that Galectin-3 might be used as a new prognostic and therapeutic biomarker in Idiopathic Parkinson's Disease (IPD). METHOD In this prospective and cross-sectional study, the Unified Parkinson's Disease Rating Scale (UPDRS) and Modified Hoehn and Yahr (H&Y) scales were applied to each patient diagnosed as IPD according to the UK Brain Bank diagnostic criteria. The control group consisted of healthy individuals with the same age, gender, and body mass index characteristics as the patients meeting the exclusion criteria. RESULTS A total of 111 cases were included in the study, 48 were IPD, and 63 were healthy controls. There were no statistically significant differences between the IPD and control groups in terms of demographic, anthropometric, and blood parameters (p > 0.05). Serum galectin-3 levels were significantly higher in IPD than the control group (p < 0.001). Serum galectin-3 levels, UPDRS scores, and duration of disease were significantly higher in patients with IPD in parallel with the progression of the disease (p < 0.001; 0.001; 0.009). No significant relationship was detected between the stage of the disease and other parameters (p < 0.05). CONCLUSION Our study supports the hypothesis that serum galectin-3 level might be associated with IPD. Our data suggest that serum galectin-3 levels might be an accessible biomarker for the detection and prevention of chronic, progressive diseases such as IPH.
Collapse
Affiliation(s)
- Hülya Olgun Yazar
- Ordu University Training and Research Hospital, Clinic of Neurology, Turkey.
| | - Tamer Yazar
- Ordu State Hospital, Clinic of Neurology, Turkey
| | - Murat Cihan
- Ordu University Training and Research Hospital, Clinical Biochemist, Turkey
| |
Collapse
|
18
|
Thomas L, Pasquini LA. Galectin-3-Mediated Glial Crosstalk Drives Oligodendrocyte Differentiation and (Re)myelination. Front Cell Neurosci 2018; 12:297. [PMID: 30258354 PMCID: PMC6143789 DOI: 10.3389/fncel.2018.00297] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 08/17/2018] [Indexed: 12/17/2022] Open
Abstract
Galectin-3 (Gal-3) is the only chimeric protein in the galectin family. Gal-3 structure comprises unusual tandem repeats of proline and glycine-rich short stretches bound to a carbohydrate-recognition domain (CRD). The present review summarizes Gal-3 functions in the extracellular and intracellular space, its regulation and its internalization and secretion, with a focus on the current knowledge of Gal-3 role in central nervous system (CNS) health and disease, particularly oligodendrocyte (OLG) differentiation, myelination and remyelination in experimental models of multiple sclerosis (MS). During myelination, microglia-expressed Gal-3 promotes OLG differentiation by binding glycoconjugates present only on the cell surface of OLG precursor cells (OPC). During remyelination, microglia-expressed Gal-3 favors an M2 microglial phenotype, hence fostering myelin debris phagocytosis through TREM-2b phagocytic receptor and OLG differentiation. Gal-3 is necessary for myelin integrity and function, as evidenced by myelin ultrastructural and behavioral studies from LGALS3-/- mice. Mechanistically, Gal-3 enhances actin assembly and reduces Erk 1/2 activation, leading to early OLG branching. Gal-3 later induces Akt activation and increases MBP expression, promoting gelsolin release and actin disassembly and thus regulating OLG final differentiation. Altogether, findings indicate that Gal-3 mediates the glial crosstalk driving OLG differentiation and (re)myelination and may be regarded as a target in the design of future therapies for a variety of demyelinating diseases.
Collapse
Affiliation(s)
- Laura Thomas
- Department of Biological Chemistry, School of Pharmacy and Biochemistry, University of Buenos Aires, Buenos Aires, Argentina.,Institute of Chemistry and Biological Physicochemistry (IQUIFIB), National Scientific and Technical Research Council (CONICET), Buenos Aires, Argentina
| | - Laura Andrea Pasquini
- Department of Biological Chemistry, School of Pharmacy and Biochemistry, University of Buenos Aires, Buenos Aires, Argentina.,Institute of Chemistry and Biological Physicochemistry (IQUIFIB), National Scientific and Technical Research Council (CONICET), Buenos Aires, Argentina
| |
Collapse
|
19
|
Siew JJ, Chern Y. Microglial Lectins in Health and Neurological Diseases. Front Mol Neurosci 2018; 11:158. [PMID: 29867350 PMCID: PMC5960708 DOI: 10.3389/fnmol.2018.00158] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2018] [Accepted: 04/25/2018] [Indexed: 12/11/2022] Open
Abstract
Microglia are the innate sentinels of the central nervous system (CNS) and are responsible for the homeostasis and immune defense of the CNS. Under the influence of the local environment and cell-cell interaction, microglia exhibit a multidimensional and context-dependent phenotypes that can be cytotoxic and neuroprotective. Recent studies suggest that microglia express multitudinous types of lectins, including galectins, Siglecs, mannose-binding lectins (MBLs) and other glycan binding proteins. Because most studies that examine lectins focus on the peripheral system, the functions of lectins have not been critically investigated in the CNS. In addition, the types of brain cells that contribute to the altered levels of lectins present in diseases are often unclear. In this review, we will discuss how galectins, Siglecs, selectins and MBLs contribute to the dynamic functions of microglia. The interacting ligands of these lectins are complex glycoconjugates, which consist of glycoproteins and glycolipids that are expressed on microglia or surrounding cells. The current understanding of the heterogeneity and functions of glycans in the brain is limited. Galectins are a group of pleotropic proteins that recognize both β-galactoside-containing glycans and non- β-galactoside-containing proteins. The function and regulation of galectins have been implicated in immunomodulation, neuroinflammation, apoptosis, phagocytosis and oxidative bursts. Most Siglecs are expressed at a low level on the plasma membrane and bind to sialic acid residues for immunosurveillance and cell-cell communication. Siglecs are classified based on their inhibitory and activatory downstream signaling properties. Inhibitory Siglecs negatively regulate microglia activation upon recognizing the intact sialic acid patterns and vice versa. MBLs are expressed upon infection in cytoplasm and can be secreted in order to recognize molecules containing terminal mannose as an innate immune defense machinery. Most importantly, multiple studies have reported dysregulation of lectins in neurological disorders. Here, we reviewed recent studies on microglial lectins and their functions in CNS health and disease, and suggest that these lectin families are novel, potent therapeutic targets for neurological diseases.
Collapse
Affiliation(s)
- Jian Jing Siew
- Taiwan International Graduate Program in Molecular Medicine, National Yang-Ming University and Academia Sinica, Taipei, Taiwan.,Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Yijuang Chern
- Taiwan International Graduate Program in Molecular Medicine, National Yang-Ming University and Academia Sinica, Taipei, Taiwan.,Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| |
Collapse
|
20
|
Cheng X, Svensson M, Yang Y, Deierborg T, Ekblad E, Voss U. Focal, but not global, cerebral ischaemia causes loss of myenteric neurons and upregulation of vasoactive intestinal peptide in mouse ileum. Int J Exp Pathol 2018; 99:38-45. [PMID: 29577471 DOI: 10.1111/iep.12263] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Accepted: 01/14/2018] [Indexed: 02/06/2023] Open
Abstract
Reduced blood flow to the brain induces cerebral ischaemia, potentially causing central injury and peripheral complications including gastrointestinal (GI) dysfunction. The pathophysiology behind GI symptoms is suspected to be neuropathy in the enteric nervous system (ENS), which is essential in regulating GI function. This study investigates if enteric neuropathy occurs after cerebral ischaemia, by analysing neuronal survival and relative numbers of vasoactive intestinal peptide (VIP) and neuronal nitric oxide synthase (nNOS) expressing neurons in mouse ileum after three types of cerebral ischaemia. Focal cerebral ischaemia, modelled by permanent middle cerebral artery occlusion (pMCAO) and global cerebral ischaemia, modelled with either transient occlusion of both common carotid arteries followed by reperfusion (GCIR) or chronic cerebral hypoperfusion (CCH) was performed on C56BL/6 mice. Sham-operated mice for each ischaemia model served as control. Ileum was collected after 1-17 weeks, depending on model, and analysed using morphometry and immunocytochemistry. For each group, intestinal mucosa and muscle layer thicknesses, neuronal numbers and relative proportions of neurons immunoreactive (IR) for nNOS or VIP were estimated. No alterations in mucosa or muscle layer thicknesses were noted in any of the groups. Loss of myenteric neurons and an increased number of VIP-IR submucous neurons were found in mouse ileum 7 days after pMCAO. None of the global ischaemia models showed any alterations in neuronal survival or relative numbers of VIP- and nNOS-IR neurons. We conclude that focal cerebral ischaemia and global cerebral ischaemia influence enteric neuronal survival differently. This is suggested to reflect differences in peripheral neuro-immune responses.
Collapse
Affiliation(s)
- Xiaowen Cheng
- Neurogastroenterology, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Martina Svensson
- Neuroinflammation Units, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Yiyi Yang
- Neuroinflammation Units, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Tomas Deierborg
- Neuroinflammation Units, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Eva Ekblad
- Neurogastroenterology, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Ulrikke Voss
- Neurogastroenterology, Department of Experimental Medical Science, Lund University, Lund, Sweden
| |
Collapse
|
21
|
Sciacchitano S, Lavra L, Morgante A, Ulivieri A, Magi F, De Francesco GP, Bellotti C, Salehi LB, Ricci A. Galectin-3: One Molecule for an Alphabet of Diseases, from A to Z. Int J Mol Sci 2018; 19:ijms19020379. [PMID: 29373564 PMCID: PMC5855601 DOI: 10.3390/ijms19020379] [Citation(s) in RCA: 249] [Impact Index Per Article: 35.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2017] [Revised: 01/18/2018] [Accepted: 01/22/2018] [Indexed: 02/07/2023] Open
Abstract
Galectin-3 (Gal-3) regulates basic cellular functions such as cell-cell and cell-matrix interactions, growth, proliferation, differentiation, and inflammation. It is not surprising, therefore, that this protein is involved in the pathogenesis of many relevant human diseases, including cancer, fibrosis, chronic inflammation and scarring affecting many different tissues. The papers published in the literature have progressively increased in number during the last decades, testifying the great interest given to this protein by numerous researchers involved in many different clinical contexts. Considering the crucial role exerted by Gal-3 in many different clinical conditions, Gal-3 is emerging as a new diagnostic, prognostic biomarker and as a new promising therapeutic target. The current review aims to extensively examine the studies published so far on the role of Gal-3 in all the clinical conditions and diseases, listed in alphabetical order, where it was analyzed.
Collapse
Affiliation(s)
- Salvatore Sciacchitano
- Department of Clinical and Molecular Medicine, Sapienza University, Policlinico Umberto I, Viale Regina Elena 324, 00161 Rome, Italy.
- Laboratory of Biomedical Research, Niccolò Cusano University Foundation, Via Don Carlo Gnocchi 3, 00166 Rome, Italy.
| | - Luca Lavra
- Laboratory of Biomedical Research, Niccolò Cusano University Foundation, Via Don Carlo Gnocchi 3, 00166 Rome, Italy.
| | - Alessandra Morgante
- Laboratory of Biomedical Research, Niccolò Cusano University Foundation, Via Don Carlo Gnocchi 3, 00166 Rome, Italy.
| | - Alessandra Ulivieri
- Laboratory of Biomedical Research, Niccolò Cusano University Foundation, Via Don Carlo Gnocchi 3, 00166 Rome, Italy.
| | - Fiorenza Magi
- Laboratory of Biomedical Research, Niccolò Cusano University Foundation, Via Don Carlo Gnocchi 3, 00166 Rome, Italy.
| | - Gian Paolo De Francesco
- Department of Oncological Science, Breast Unit, St Andrea University Hospital, Via di Grottarossa, 1035/39, 00189 Rome, Italy.
| | - Carlo Bellotti
- Operative Unit Surgery of Thyroid and Parathyroid, Sapienza University of Rome, S. Andrea Hospital, Via di Grottarossa, 1035/39, 00189 Rome, Italy.
| | - Leila B Salehi
- Laboratory of Biomedical Research, Niccolò Cusano University Foundation, Via Don Carlo Gnocchi 3, 00166 Rome, Italy.
- Department of Biopathology and Diagnostic Imaging, Tor Vergata University, Via Montpellier 1, 00133 Rome, Italy.
| | - Alberto Ricci
- Department of Clinical and Molecular Medicine, Sapienza University, Policlinico Umberto I, Viale Regina Elena 324, 00161 Rome, Italy.
| |
Collapse
|
22
|
Boziki M, Polyzos SA, Deretzi G, Kazakos E, Katsinelos P, Doulberis M, Kotronis G, Giartza-Taxidou E, Laskaridis L, Tzivras D, Vardaka E, Kountouras C, Grigoriadis N, Thomann R, Kountouras J. A potential impact of Helicobacter pylori-related galectin-3 in neurodegeneration. Neurochem Int 2017; 113:137-151. [PMID: 29246761 DOI: 10.1016/j.neuint.2017.12.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Revised: 12/03/2017] [Accepted: 12/11/2017] [Indexed: 02/07/2023]
Abstract
Neurodegeneration represents a component of the central nervous system (CNS) diseases pathogenesis, either as a disability primary source in the frame of prototype neurodegenerative disorders, or as a secondary effect, following inflammation, hypoxia or neurotoxicity. Galectins are members of the lectin superfamily, a group of endogenous glycan-binding proteins, able to interact with glycosylated receptors expressed by several immune cell types. Glycan-lectin interactions play critical roles in the living systems by involving and mediating a variety of biologically important normal and pathological processes, including cell-cell signaling shaping cell communication, proliferation and migration, immune responses and fertilization, host-pathogen interactions and diseases such as neurodegenerative disorders and tumors. This review focuses in the role of Galectin-3 in shaping responses of the immune system against microbial agents, and concretely, Helicobacter pylori (Hp), thereby potentiating effect of the microbe in areas distant from the ordinary site of colonization, like the CNS. We hereby postulate that gastrointestinal Hp alterations in terms of immune cell functional phenotype, cytokine and chemokine secretion, may trigger systemic responses, thereby conferring implications for remote processes susceptible in immunity disequilibrium, namely, the CNS inflammation and/or neurodegeneration.
Collapse
Affiliation(s)
- Marina Boziki
- Department of Medicine, Second Medical Clinic, Aristotle University of Thessaloniki, Ippokration Hospital, Thessaloniki, Greece; Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Stergios A Polyzos
- Department of Medicine, Second Medical Clinic, Aristotle University of Thessaloniki, Ippokration Hospital, Thessaloniki, Greece
| | - Georgia Deretzi
- Department of Neurology, Multiple Sclerosis Unit, Papageorgiou Hospital, Thessaloniki, Greece
| | - Evangelos Kazakos
- Department of Medicine, Second Medical Clinic, Aristotle University of Thessaloniki, Ippokration Hospital, Thessaloniki, Greece
| | - Panagiotis Katsinelos
- Department of Medicine, Second Medical Clinic, Aristotle University of Thessaloniki, Ippokration Hospital, Thessaloniki, Greece
| | - Michael Doulberis
- Department of Medicine, Second Medical Clinic, Aristotle University of Thessaloniki, Ippokration Hospital, Thessaloniki, Greece; Department of Internal Medicine, Bürgerspital Solothurn, Solothurn, Switzerland
| | - Georgios Kotronis
- Department of Medicine, Second Medical Clinic, Aristotle University of Thessaloniki, Ippokration Hospital, Thessaloniki, Greece
| | - Evaggelia Giartza-Taxidou
- Department of Medicine, Second Medical Clinic, Aristotle University of Thessaloniki, Ippokration Hospital, Thessaloniki, Greece
| | - Leonidas Laskaridis
- Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Dimitri Tzivras
- Department of Medicine, Second Medical Clinic, Aristotle University of Thessaloniki, Ippokration Hospital, Thessaloniki, Greece
| | - Elisabeth Vardaka
- Department of Medicine, Second Medical Clinic, Aristotle University of Thessaloniki, Ippokration Hospital, Thessaloniki, Greece
| | - Constantinos Kountouras
- Department of Medicine, Second Medical Clinic, Aristotle University of Thessaloniki, Ippokration Hospital, Thessaloniki, Greece
| | - Nikolaos Grigoriadis
- Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Robert Thomann
- Department of Internal Medicine, Bürgerspital Solothurn, Solothurn, Switzerland
| | - Jannis Kountouras
- Department of Medicine, Second Medical Clinic, Aristotle University of Thessaloniki, Ippokration Hospital, Thessaloniki, Greece.
| |
Collapse
|
23
|
Ohlsson B, Gustafsson R, Swahn F, Toth E, Veress B, Thorlacius H. Endoscopic full-thickness biopsy, a novel method in the work up of complicated abdominal symptoms. Therap Adv Gastroenterol 2017; 11:1756283X17730747. [PMID: 29383022 PMCID: PMC5784575 DOI: 10.1177/1756283x17730747] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Gastrointestinal complaints without obvious organic causes confirmed by clinical laboratory analyses, endoscopy or radiology are often referred to functional entities. Irritable bowel syndrome (IBS) is the most common functional disorder in the gut. Careful examination of these patients may reveal other diagnoses of defined etiologies, e.g., enteric neuropathy, microscopic colitis, and primary Sjögre's syndrome. The present case describes a young patient with incapacitating gastrointestinal symptoms presumed to be IBS, who underwent endoscopic full-thickness biopsy in sigmoid colon. Histopathological examination revealed degenerative enteric neuropathy, possibly secondary to chronic ischemia.
Collapse
Affiliation(s)
- Bodil Ohlsson
- Department of Clinical Sciences, Lund University, Skåne University Hospital, Jan Waldenström Street 15, S-205 02 Malmö, Sweden
| | - Rita Gustafsson
- Department of Gastroenterology, Skane University Hospital, Malmö, Sweden
| | - Fredrik Swahn
- Department of Surgery, Lund University, Skane University Hospital, Lund, Sweden
| | - Ervin Toth
- Department of Gastroenterology, Skane University Hospital, Malmö, Sweden
| | - Béla Veress
- Department of Cellular Pathology, Unilabs AB, Skövde, Sweden
| | - Henrik Thorlacius
- Department of Surgery, Lund University, Skane University Hospital, Malmö, Sweden
| |
Collapse
|
24
|
Abstract
Microbiota research, in particular that of the gut, has recently gained much attention in medical research owing to technological advances in metagenomics and metabolomics. Despite this, much of the research direction has focused on long-term or chronic effects of microbiota manipulation on health and disease. In this addendum, we reflect on our recent publication that reported findings addressing a rather unconventional hypothesis. Bacterial pneumonia is highly prevalent and is one of the leading contributors to stroke morbidity and mortality worldwide. However, microbiological cultures of samples taken from stroke patient with a suspected case of pneumonia often return with a negative result. Therefore, we proposed that post-stroke infection may be due to the presence of anaerobic bacteria, possibly those originated from the host gut microbiota. Supporting this, we showed that stroke promotes intestinal barrier breakdown and robust microbiota changes, and the subsequent translocation of selective bacterial strain from the host gut microbiota to peripheral tissues (i.e. lung) induces post-stroke infections. Our findings were further supported by various elegant studies published in the past 12 months. Here, we discuss and provide an overview of our key findings, supporting studies, and the implications for future advances in stroke research.
Collapse
Affiliation(s)
- Shu Wen Wen
- Centre for Inflammatory Diseases, Department of Medicine, School of Clinical Sciences, Monash University, Clayton, Victoria, Australia
| | - Connie H. Y. Wong
- Centre for Inflammatory Diseases, Department of Medicine, School of Clinical Sciences, Monash University, Clayton, Victoria, Australia,CONTACT Connie H. Y. Wong, PhD. , Centre for Inflammatory Diseases, Department of Medicine, School of Clinical Sciences, Monash Medical Centre, Monash University, Clayton, VIC 3168 Australia
| |
Collapse
|