A key role of nutrition and gut microbiota in the development of neurodegenerative diseases

2016 ◽  
Vol 6 (4) ◽  
pp. 36-46
Author(s):  
A.B. Danilov ◽  
◽  
A.V. Prishchepa ◽  
2018 ◽  
Vol 19 (6) ◽  
pp. 478-489 ◽  
Author(s):  
Francesco Di Meo ◽  
Stella Donato ◽  
Alba Di Pardo ◽  
Vittorio Maglione ◽  
Stefania Filosa ◽  
...  

Pathogens ◽  
2021 ◽  
Vol 10 (7) ◽  
pp. 887
Author(s):  
Josephine Trichka ◽  
Wen-Quan Zou

The process of neuroinflammation contributes to the pathogenic mechanism of many neurodegenerative diseases. The deleterious attributes of neuroinflammation involve aberrant and uncontrolled activation of glia, which can result in damage to proximal brain parenchyma. Failure to distinguish self from non-self, as well as leukocyte reaction to aggregation and accumulation of proteins in the CNS, are the primary mechanisms by which neuroinflammation is initiated. While processes local to the CNS may instigate neurodegenerative disease, the existence or dysregulation of systemic homeostasis can also serve to improve or worsen CNS pathologies, respectively. One fundamental component of systemic homeostasis is the gut microbiota, which communicates with the CNS via microbial metabolite production, the peripheral nervous system, and regulation of tryptophan metabolism. Over the past 10–15 years, research focused on the microbiota–gut–brain axis has culminated in the discovery that dysbiosis, or an imbalance between commensal and pathogenic gut bacteria, can promote CNS pathologies. Conversely, a properly regulated and well-balanced microbiome supports CNS homeostasis and reduces the incidence and extent of pathogenic neuroinflammation. This review will discuss the role of the gut microbiota in exacerbating or alleviating neuroinflammation in neurodegenerative diseases, and potential microbiota-based therapeutic approaches to reduce pathology in diseased states.


2020 ◽  
Vol 21 ◽  
Author(s):  
Tingting Zhao ◽  
Zhe Wang ◽  
Zhilong Liu ◽  
Youhua Xu

: With the recognition of the important role of gut microbiota in both health and disease progression, attempts to modulate its composition as well as its co-metabolism with the organism have attracted special attention. Abundant studies have demonstrated that dysfunction or imbalance of gut microbiota is closely with disease including endocrine diseases, neurodegenerative diseases, tumors, cardiovascular diseases, et al. Herbal medicines have been applied for preventing and treating disease worldwide for hundreds of years. Although the underlying mechanism seems to be complex, one of the important one is through modulating gut microbiota. In this review, co-metabolism between herbal medicines and microbiota, as well as the potential pathways are summarized from most recent published papers.


Nutrients ◽  
2021 ◽  
Vol 13 (9) ◽  
pp. 2940
Author(s):  
Diogo Carregosa ◽  
Sara Mota ◽  
Sofia Ferreira ◽  
Beatriz Alves-Dias ◽  
Natasa Loncarevic-Vasiljkovic ◽  
...  

The rise of neurodegenerative diseases in an aging population is an increasing problem of health, social and economic consequences. Epidemiological and intervention studies have demonstrated that diets rich in (poly)phenols can have potent health benefits on cognitive decline and neurodegenerative diseases. Meanwhile, the role of gut microbiota is ever more evident in modulating the catabolism of (poly)phenols to dozens of low molecular weight (poly)phenol metabolites that have been identified in plasma and urine. These metabolites can reach circulation in higher concentrations than parent (poly)phenols and persist for longer periods of time. However, studies addressing their potential brain effects are still lacking. In this review, we will discuss different model organisms that have been used to study how low molecular weight (poly)phenol metabolites affect neuronal related mechanisms gathering critical insight on their potential to tackle the major hallmarks of neurodegeneration.


Antioxidants ◽  
2021 ◽  
Vol 10 (9) ◽  
pp. 1370
Author(s):  
Umair Shabbir ◽  
Akanksha Tyagi ◽  
Fazle Elahi ◽  
Simon Okomo Aloo ◽  
Deog-Hwan Oh

Gut microbiota (GM) play a role in the metabolic health, gut eubiosis, nutrition, and physiology of humans. They are also involved in the regulation of inflammation, oxidative stress, immune responses, central and peripheral neurotransmission. Aging and unhealthy dietary patterns, along with oxidative and inflammatory responses due to gut dysbiosis, can lead to the pathogenesis of neurodegenerative diseases, especially Alzheimer’s disease (AD). Although the exact mechanism between AD and GM dysbiosis is still unknown, recent studies claim that secretions from the gut can enhance hallmarks of AD by disturbing the intestinal permeability and blood–brain barrier via the microbiota–gut–brain axis. Dietary polyphenols are the secondary metabolites of plants that possess anti-oxidative and anti-inflammatory properties and can ameliorate gut dysbiosis by enhancing the abundance of beneficial bacteria. Thus, modulation of gut by polyphenols can prevent and treat AD and other neurodegenerative diseases. This review summarizes the role of oxidative stress, inflammation, and GM in AD. Further, it provides an overview on the ability of polyphenols to modulate gut dysbiosis, oxidative stress, and inflammation against AD.


2021 ◽  
Vol 41 ◽  
pp. 01002
Author(s):  
Daria Guseva

The link between nutrition and human diseases has always been recognized, but only with modern molecular genetic analysis tools, the role of the gut microbiome including gastrointestinal tract function on neurodegenerative diseases has become obvious. Gut microbiota significantly influences metabolic and immune responses of the host organism, and thus, dietor infection-related imbalances (dysbiosis) of the gut microbiota disrupt the local and systemic homeostasis and often lead to digestive diseases, such as inflammatory bowel diseases (IBD), type II diabetes, obesity, as well as neurological diseases. The association of the gastrointestinal tract diseases with neurodegenerative diseases, as well as mental and neurological disorders such as depression, anxiety, autism, and schizophrenia has been described. This interaction is called the gut-brain axis and represents one of the most relevant targets for the treatment of IBD and cardiovascular and neurological diseases. One of the most important regulators of this axis is the gut microbiota and its metabolites, which influence brain functions via the immune system, tryptophan metabolism, vagus nerve, and enteric nervous system. We are studying the importance of gut microbiota and their metabolites in immune and glial/microglial reaction in the gastrointestinal tract and central nervous system, to provide valuable insights into the functional potential of the microbiome on the gut-brain axis via specific metabolites. We combine several modern techniques, including molecular manipulations, advanced cellular imaging techniques, and behavioral approaches, to address additional systems-level questions.


2019 ◽  
Vol 20 (19) ◽  
pp. 4704 ◽  
Author(s):  
Valeria D’Argenio ◽  
Daniela Sarnataro

Misfolded and abnormal β-sheets forms of wild-type proteins, such as cellular prion protein (PrPC) and amyloid beta (Aβ), are believed to be the vectors of neurodegenerative diseases, prion and Alzheimer’s disease (AD), respectively. Increasing evidence highlights the “prion-like” seeding of protein aggregates as a mechanism for pathological spread in AD, tauopathy, as well as in other neurodegenerative diseases, such as Parkinson’s. Mutations in both PrPC and Aβ precursor protein (APP), have been associated with the pathogenesis of these fatal disorders with clear evidence for their pathogenic significance. In addition, a critical role for the gut microbiota is emerging; indeed, as a consequence of gut–brain axis alterations, the gut microbiota has been involved in the regulation of Aβ production in AD and, through the microglial inflammation, in the amyloid fibril formation, in prion diseases. Here, we aim to review the role of microbiome (“the other human genome”) alterations in AD and prion disease pathogenesis.


2020 ◽  
Vol 8 (5) ◽  
pp. 784 ◽  
Author(s):  
Mark Obrenovich ◽  
Hayden Jaworski ◽  
Tara Tadimalla ◽  
Adil Mistry ◽  
Lorraine Sykes ◽  
...  

The human gut hosts a wide and diverse ecosystem of microorganisms termed the microbiota, which line the walls of the digestive tract and colon where they co-metabolize digestible and indigestible food to contribute a plethora of biochemical compounds with diverse biological functions. The influence gut microbes have on neurological processes is largely yet unexplored. However, recent data regarding the so-called leaky gut, leaky brain syndrome suggests a potential link between the gut microbiota, inflammation and host co-metabolism that may affect neuropathology both locally and distally from sites where microorganisms are found. The focus of this manuscript is to draw connection between the microbiota–gut–brain (MGB) axis, antibiotics and the use of “BUGS AS DRUGS” for neurodegenerative diseases, their treatment, diagnoses and management and to compare the effect of current and past pharmaceuticals and antibiotics for alternative mechanisms of action for brain and neuronal disorders, such as Alzheimer disease (AD), Amyotrophic Lateral Sclerosis (ALS), mood disorders, schizophrenia, autism spectrum disorders and others. It is a paradigm shift to suggest these diseases can be largely affected by unknown aspects of the microbiota. Therefore, a future exists for applying microbial, chemobiotic and chemotherapeutic approaches to enhance translational and personalized medical outcomes. Microbial modifying applications, such as CRISPR technology and recombinant DNA technology, among others, echo a theme in shifting paradigms, which involve the gut microbiota (GM) and mycobiota and will lead to potential gut-driven treatments for refractory neurologic diseases.


Sign in / Sign up

Export Citation Format

Share Document