Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 + 2316 word(s) 2316 2021-07-01 05:56:53 |
2 corrected the format Meta information modification 2316 2021-07-14 10:48:35 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Rai, S.N. Neurodegenerative Pathogenesis. Encyclopedia. Available online: https://encyclopedia.pub/entry/12065 (accessed on 29 March 2024).
Rai SN. Neurodegenerative Pathogenesis. Encyclopedia. Available at: https://encyclopedia.pub/entry/12065. Accessed March 29, 2024.
Rai, Sachchida Nand. "Neurodegenerative Pathogenesis" Encyclopedia, https://encyclopedia.pub/entry/12065 (accessed March 29, 2024).
Rai, S.N. (2021, July 14). Neurodegenerative Pathogenesis. In Encyclopedia. https://encyclopedia.pub/entry/12065
Rai, Sachchida Nand. "Neurodegenerative Pathogenesis." Encyclopedia. Web. 14 July, 2021.
Neurodegenerative Pathogenesis
Edit

Current research in medicine in several parts of the world has attempted to establish a link between the occurrence of neurodegenerative pathologies, microbiota dysbiosis, and the incidence of obesity. The body’s response to different physicochemical factors has also been influenced by the proper assimilation of bioactive compounds contained in the food that is ingested. Oxidative stress is one of the major factors that directly affects the functioning of the human microbiota.

gut-brain axis neurotransmitter dysbiosis obesity

1. Introduction

Recovery from dysbiosis (the imbalance in gut microbial population) and the establishment of eubiosis are currently being investigated in research regarding a demonstration of a direct relationship between the nervous system and the colon’s microbial fingerprint [1]. Considered to be the second brain (enteric nervous system), the structure of enteric neurons is the key factor that controls the physiological response to the environment [2]. Increased administration of antibiotics determines the prevalence of antibiotic resistance genes in gut bacterial strains, and sustains dysbiosis [3]. Molecular interactions between microbial strains play an essential role in the stress response, in developing pathologies, and in reducing inflammatory progression, and are a significant factor in supporting and maintaining dysbiosis [4].

Oxidative stress represents one major factor that directly affects the structure (microbial pattern) of human microbiota [5]. The perturbed pattern determines the incidence of degenerative diseases, obesity, and other physiological modifications [6].

The regenerative (modulatory) function of microbiota has been recently demonstrated in research to reduce significant pathology symptoms, mainly those involved in degenerative diseases [7]. This aspect results from microbiota-targeted therapies that modulate the microbial pattern and influence the host homeostasis [8]. Restoring the microbiota will lead to better overall functioning, which also helps maintain the functionality of other tissues or organs [7].

This evidence linking gut microbiota to degenerative pathologies is known, and new links between these are needed in the current epidemiological context. A newly identified aspect was obesity, based on its connection with the microbiota’s role. Identifying critical mediators of this process is a novelty that will open up the possibility of new therapeutic approaches by correcting dysbiosis and reducing the inflammatory response.

From clinical trials in patients with neurodegenerative diseases, the effect of these products has been unclear. Positive results tend to be the improvement of other physiological parameters (anxiety reduction or biochemical parameters [9]) that act indirectly on the cognitive status [10]. SCFAs were just key mediators, which modulate the colon’s integrity. The initial stages are developed, in many cases, at the level of the nervous system by its improper function that influences gut microbiota status.

2. Microbiota Role in the General State of Health

These regenerative aspects are closely related to inflammatory progression, as microbial profile dynamics can ensure host homeostasis. A clear picture could be represented by microbial species’ differences to observe the microbial balance in various chronic pathologies. An example of correspondence between the microbial population of microbiota, obesity, and neurodegenerative pathologies could be the variation ofAkkermansia muciniphilapopulation [11]:In obesity, there was an increase in theFirmicutes:Bacteroidetesratio (number of the cells/mL) The differences in microbial composition determined the increased abundance of pathways involved in alpha-linolenic acid metabolism, electron transfer carriers, bacterial motility proteins, Parkinson’s disease, and Prion diseases [12].

The colonic microbiota activity and the metabolic function’s modulation depend on bioactive compounds’ bioavailability [13]. Modulation by functional products influences both the microbial fingerprint, which has a xenobiotic effect, and the products of microbial metabolism. Reducing the xenobiotic impact is a complex process that affects the regenerative function [14]. The functioning of these two systems is based on an interdependence that expresses a capacity (the physiological barrier’s function) to maintain an optimal state of health [15].

3. Oxidative Stress and Microbiota in Neurodegenerative Processes

Maintaining a well balanced relationship between intestinal microbiota and the nervous system is essential in promoting homeostasis [16]. The onset of dysbiosis influences the occurrence of cognitive disorders (depression, anxiety, and associated dysfunction, such as multiple sclerosis [17]) that coincide with the onset and establishment of irritability, sensitivity, and minor neurological diseases [18]. These manifestations are sustained by inflammatory processes that characterize intestinal dysbiosis [19]. Modulation of the microbial pattern eliminates these symptoms (as multitarget strategy) [20], and classical medication of neurodegenerative pathology as AD only determines an improvement of cognitive status [21].

Inflammatory progression and aggravation reduce the natural process of neuronal protection [22], resulting in premature neuronal aging. This phenomenon frequently leads to the establishment of neurodegeneration [23]; modulation of the microbiota contributes to improving these complex processes and reducing accumulations of proinflammatory factors [7].

Nutraceuticals can be the key to this process, which controls the overall health status with long-term effects [24]. The control of these functions is also a way of reducing the natural aging process ’s impact strictly related to cell oxidation and the action of oxidative stress [25]. This aspect demonstrated an imbalance between free radicals and the human body’s

Thus, antioxidants directly attenuate biological factors that can cause inflammatory progression [26]. The use of classical drugs, on the other hand, may generate toxicity and adverse reactions. Antioxidant products reduce this toxicity by the scavenging activities of free radicals. These compounds, which are often polyphenol carboxylic compounds, have other effects and, after administration, are identified as prebiotic-like products [27].

The complex relationship between biotransformation at the microbiota level and the protection against neurodegeneration results in reducing oxidative stress (an increase in antioxidant protection) and preventing the aging of nerve cells. Changing bowel motility, increased intestinal permeability, and increased drug consumption with side effects is another essential factor that correlates (or absorptive dysfunction represents) critical factors. They associate the natural aging process with a reduced immune response and an increased inflammatory progression. The interconnections between physiological processes associated with old age progressively lead to the establishment of the neurodegeneration process [1].

The regenerative function is expressed at two levels:Modulation (correction) of the disturbing pattern (dysbiosis);Modulation (correction) of physiological processes resulting from restoring the balance along the gut–brain axis.

Even if the microbiota’s regenerative role is not widely accepted in clinical practice, its unbalanced metabolism favors the onset of the pathological state where oxidative stress has a more pronounced character (such as in the cardiovascular or neuronal systems) [28]. The critical point is the establishment of eubiosis, which could influence both mechanisms [29]. Metabolic correction provides a biochemical explanation for optimizing physiological functions, and the oxidative cause of stress can be eliminated [30].

Clinical relevance was determined by exo- and endogenous factors. The modulators’ chemical structure is an essential element that influences the gut-brain (biochemical signaling) balance [31]. Disruption of two-way communication is investigated in recent research, the source of neurological dysfunctions, and the cause of a decreased body immune response [32].

Microbiota activity can be controlled through the gut-brain axis using different compounds with particular functionalities based on several pathways [33] called neurotransmitters; for example, identifying the neurotransmitter pattern’s role is crucial in explaining the interaction with different exogenous factors [28][34][35].

Neuroendocrine systems also interact with the gut microbiome, controlling physiological functions in response to oxidative stress by the hypothalamic-pituitary-adrenal (HPA) axis. As different microbiota-gut-brain axis pathologies are associated with the HPA axis dysfunction [36], this interaction is significant. In PD, the triggering of limb tremors disrupts dopamine synthesis, which controls and regulates muscle movements through the gut–brain axis. The phenomenon is linked to the microbiota pattern; therefore, the microbial profile’s control determines a secondary (indirect) regulation of dopamine synthesis [37].

Recent studies have shown that microbiota plays a significant role in the pathogenesis and therapy of PD [37]. The dysbiosis of microbiota may indicate the progression of PD [38]. Several patients with PD exhibit dysbiosis and gastrointestinal problems even before the onset of motor symptoms and their diagnosis [39]. Before the clinical onset of PD symptoms, leaky gut was observed due to gastrointestinal symptoms like constipation, hypersalivation, and dysphagia [38].

The population of pathogenic bacteria like Enterobacteria, Streptococci, Staphylococci,Shigella, andH. pyloriwas also increased in PD cases [40][41]. Gut microbes affect the synthesis of dopamine [42], deposition of α-synuclein enhances oxidative stress, induces local inflammation, enhances intestinal permeability, and causes constipation in PD [43][44][45]. ; therefore, gut microbiota can be explored for the early diagnosis of PD, as suggested by current studies, which are very reliable and noninvasive [46]. In PD,Helicobacter pyloriare mentioned, with increasing levels of Enterobacteriaceae and a lower number ofPrevotellaceae[45].

The Mediterranean diet can also sustain a healthy microbiome pattern and reduce the risk of PD by enhancing the growth of beneficial strains [39][47]; however, several side effects may be mentioned at a preprolonged administration of these functional products: intestinal bacterial overgrowth, D-lactate acidosis, brain fogginess, and horizontal gene transfer [48]. The dysbiosis of microbiota may represent an early sign of PD by identifying the microbiota pattern. The gastrointestinal tract initiates a spread of α-Syn, which means that initial nonmotor symptoms like constipation might be an early sign of the disease. Several probiotics, prebiotics, and their combination might restore healthy microbiota patterns and prevent further progression of PD [37][39][49].

In AD, neurotransmitters’ activity that influences neuroinflammation directly relates to the metabolic activity [50]. [50][51], and are located, especially, around the essential internal organs (e.g., liver or heart) [52]. Microbiota activity influences the formation of amyloid plaque, which progressively causes the death of neuronal cells [53]. In vivo studies have shown a decrease in certain bacteria such asEubacterium rectaleandBacteroides fragilisand an increase inEscherichia coli[54].

Other compounds with a negative effect are proinflammatory cytokines that cause severe inflammatory responses [55]. Increased synthesis of proinflammatory cytokines such as TNF-α, IFN-γ, IL-1β, IL-6, and IL-18 has been identified in people diagnosed with AD. Of these, two proinflammatory cytokines (TNF and IL-1β) were determined to have elevated values demonstrating the severity of AD [56]. The presence of these cytokines is correlated with the loss of the ratio betweenE. coliandE. rectale(the ratio is different between target groups, daily habits, etc.), which supports a typical microbial pattern in the case of degenerative diseases [57].

Amyotrophic lateral sclerosis (ALS) determines changes in the microbiota structure and dysfunction of the gut-brain axis [58]. It has been demonstrated thatAkkermansia muciniphilareduces ALS symptoms by accumulating nicotinamides that improve motor function. An inverse effect was associated with the presence ofRuminococcus torquesandParabacteroides distasonis[59].

Based on this knowledge, we could consider that modulation of microbiota could sustain neuronal health as an alternative strategy for the population’s target group. Unbalanced metabolism favors the onset of the pathological state where oxidative stress has a more pronounced character (such as in the cardiovascular or neuronal systems). The control through the gut-brain axis involves several molecules from different pathways that do not have a well described mechanism. It is necessary to identify new biomarkers and therapeutic strategies for early diagnosis and innovative treatments, especially in preventing neurodegenerative diseases.

Food addiction and drug use have a negative influence, leading to a disturbance of the neural system’s functionality. This behavior, doubled by the high degree of food processing, leads to obesity and other degenerative pathologies [60]. The initiation of dysbiosis, a result of fast-food consumption, is related to disruption in dopamine release [61][62].

Recent studies have shown a connection between obesity, type 2 diabetes, and neurodegenerative diseases. Modulation of the microbiota associated with weight loss is among the new therapeutic strategies against neurodegenerative disorders [63]. Obesity is a risk factor in developing neurodegenerative diseases (e.g., AD or PD), with common causes such as oxidative stress and mitochondrial dysfunction, both of which are supported by inflammatory progression. From a molecular perspective, early aging causes an increase in the blood-brain barrier’s permeability and intestinal permeability in the elderly, which has a proinflammatory effect [64][65].

The relationship between different bacterial strains and a dysbiotic microbiome state responds to exogenous factors and inflammatory processes generated by oxidative stress inE. colihave been reported in overweight women [66]. These data are correlated with recurrent infections caused by this strain in the urinary tract in people diagnosed with type 2 diabetes who are also overweight [67]. A reduction in oxygen consumption and overexpression of proinflammatory mediator genes (e.g., IL-6) has been demonstrated [5].

Such processes induce oxidative stress, which controls several genes’ expression including those responsible for producing various cytokines [37]. The cellular level of ROS conditions the physiological response that controls inflammatory processes. For neurodegenerative diseases (e.g., AD), redox balance is a crucial factor, andLactobacillusstrains play an essential role in maintaining the gut–brain axis homeostasis. The different physiological dysfunction (degenerative process) is closely linked to pathogenesis of inflammatory diseases (e.g., obesity or inflammatory bowel disease)

The host homeostasis was influenced not only by the level of SCFA but through other cellular components. LPS are toll-like receptors (TLR) expressed by immune cells. Maintaining intestinal health and healing the possible lesions implies microbiota-mediated TLR signals. A new approach to the influence on the microbiota may be related to the amount of lipopolysaccharides produced by bacteria (determined by the perturbed intestinal permeability), given that the amount of lipopolysaccharides in the blood was correlated with diabetes, cell death, or other diseases such as sepsis, atherosclerosis, inflammatory bowel disease, nonalcoholic fatty liver disease or neurodegenerative diseases [68][69].

Other components, such as secondary bile acids produced by microbiota action, play an essential role in controlling the production of bile acids and immunity [70]. Studies show that SCFAs and conjugated primary bile acids act differently, the first with an anti-inflammatory role, the second with the opposite role [71]. Some of these compounds act as aryl hydrocarbon receptor ligands, and their low levels are indicators of metabolic disorders. Simultaneously, through SCFAs and bile acids, gut microbiota also modulates the metabolism of tryptophan to serotonin; the final product of its degradation is another parameter in metabolic diseases [72].

The point of connection of neurodegenerative diseases (e.g., AD) with the intestinal microbiota is the microbial diversity changes that are often encountered with age. Research conducted in vitro supports these data, which indicates a dysbiotic condition in type 2 diabetes, which is related to obesity [67]. In ALS, the results were significant becauseA. muciniphilais considered part of a new type of functional product used to control weight and associated pathologies (reduce glucose intolerance) These data indicate another link between degenerative pathologies (e.g., ALS) with a decrease in the abundance of bacterial strains that help control obesity.

References

  1. Raskov, H.; Burcharth, J.; Pommergaard, H.C. Linking gut microbiota to colorectal cancer. J. Cancer. 2017, 20, 3378–3395.
  2. Mayer, E.A. Gut feelings: The emerging biology of gut-brain communication. Nat. Rev. Neurosci. 2011, 13, 453–466.
  3. Zhang, S.; Chen, D.C. Facing a new challenge: The adverse effects of antibiotics on gut microbiota and host immunity. Chin. Med. J. 2019, 132, 1135–1138.
  4. Kho, Z.Y.; Lal, S.K. The human gut microbiome–A potential controller of wellness and disease. Front. Microbiol. 2018, 9, 835.
  5. Vamanu, E. Polyphenolic nutraceuticals to combat oxidative stress through microbiota modulation. Front. Pharmacol. 2019, 10, 492.
  6. Mazon, J.N.; de Mello, A.H.; Ferreira, G.K.; Rezin, G.T. The impact of obesity on neurodegenerative diseases. Life Sci. 2017, 182, 22–28.
  7. Belkaid, Y.; Hand, T.W. Role of the microbiota in immunity and inflammation. Cell 2014, 157, 121–141.
  8. Zheng, D.; Liwinski, T.; Elinav, E. Interaction between microbiota and immunity in health and disease. Cell Res. 2020, 30, 492–506.
  9. Cheng, L.H.; Liu, Y.W.; Wu, C.C.; Wang, S.; Tsai, Y.C. Psychobiotics in mental health, neurodegenerative and neurodevelopmental disorders. J. Food Drug Anal. 2019, 27, 632–648.
  10. Den, H.; Dong, X.; Chen, M.; Zou, Z. Efficacy of probiotics on cognition, and biomarkers of inflammation and oxidative stress in adults with Alzheimer’s disease or mild cognitive impairment—A meta-analysis of randomized controlled trials. Aging 2020, 12, 4010–4039.
  11. Pleasure, D.; Guo, F.; Chechneva, O.; Bannerman, P.; Mcdonough, J.; Burns, T. Pathophysiology and treatment of canavan disease. Neurochem. Res. 2020, 45, 561–565.
  12. McGaughey, K.D.; Yilmaz-Swenson, T.; Elsayed, N.M.; Cruz, D.A.; Rodriguiz, R.M.; Kritzer, M.D.; Peterchev, A.V.; Roach, J.; Wetsel, W.C.; Williamson, D.E. Relative abundance of Akkermansia spp. and other bacterial phylotypes correlates with anxiety- and depressive-like behavior following social defeat in mice. Sci. Rep. 2019, 9, 3281.
  13. Luca, L.; Oroian, M. The impact of potential prebiotics inulin, oligofructose and potato starch on the growth of Lactobacillus casei. AgroLife Sci. J. 2019, 8, 153–159. Available online: (accessed on 19 June 2021).
  14. Koppel, N.; Rekdal, V.M.; Balskus, E.P. Chemical transformation of xenobiotics by the human gut microbiota. Science 2017, 356, eaag2770.
  15. Kotas, M.E.; Medzhitov, R. Homeostasis, inflammation, and disease susceptibility. Cell 2015, 26, 816–827.
  16. Ma, Q.; Xing, C.; Long, W.; Wang, H.Y.; Liu, Q.; Wang, R.F. Impact of microbiota on central nervous system and neurological diseases: The gut-brain axis. J. Neuroinflammation 2019, 16, 53.
  17. Feinstein, A.; Brochet, B.; Sumowski, J. The cognitive effects of anxiety and depression in immune-mediated inflammatory diseases. Neurology 2019, 92, 5.
  18. Foster, J.A.; McVey Neufeld, K.A. Gut-brain axis: How the microbiome influences anxiety and depression. Trends Neurosci. 2013, 36, 305–312.
  19. Zhu, S.; Jiang, Y.; Xu, K.; Cui, M.; Ye, W.; Zhao, G.; Jin, L.; Chen, X. The progress of gut microbiome research related to brain disorders. J. Neuroinflamm. 2020, 17, 25.
  20. Bonfili, L.; Cecarini, V.; Gogoi, O.; Gong, C.; Cuccioloni, M.; Angeletti, M.; Rossi, G.; Eleuteri, A.M. Microbiota modulation as preventative and therapeutic approach in Alzheimer’s disease. FEBS J. 2020.
  21. DeGruttola, A.K.; Low, D.; Mizoguchi, A.; Mizoguchi, E. Current understanding of dysbiosis in disease in human and animal models. Inflamm. Bowel Dis. 2016, 22, 1137–1150.
  22. Sochocka, M.; Diniz, B.S.; Leszek, J. Inflammatory response in the CNS: Friend or Foe? Mol. Neurobiol. 2017, 54, 8071–8089.
  23. Castelli, V.; Benedetti, E.; Antonosante, A.; Catanesi, M.; Pitari, G.; Ippoliti, R.; Cimini, A.; d’Angelo, M. Neuronal Cells rearrangement during aging and neurodegenerative disease: Metabolism, oxidative stress and organelles dynamic. Front. Mol. Neurosci. 2019, 12, 132.
  24. Das, L.; Bhaumik, E.; Raychaudhuri, U.; Chakraborty, R. Role of nutraceuticals in human health. J. Food Sci. Technol. 2012, 49, 173–183.
  25. Pizzino, G.; Irrera, N.; Cucinotta, M.; Pallio, G.; Mannino, F.; Arcoraci, V.; Squadrito, F.; Altavilla, D.; Bitto, A. Oxidative stress: Harms and benefits for human health. Oxid. Med. Cel. Longev. 2017, 2017, 8416763.
  26. Kurutas, E.B. The importance of antioxidants which play the role in cellular response against oxidative/nitrosative stress: Current state. Nutr. J. 2016, 15, 71.
  27. Lobo, V.; Patil, A.; Phatak, A.; Chandra, N. Free radicals, antioxidants and functional foods: Impact on human health. Pharmacog. Rev. 2010, 4, 118–126.
  28. Dumitrescu, L.; Popescu-Olaru, I.; Cozma, L.; Tulbă, D.; Hinescu, M.E.; Ceafalan, L.C.; Gherghiceanu, M.; Popescu, B.O. Oxidative Stress and the Microbiota-Gut-Brain Axis. Oxid. Med. Cel. Longev. 2018, 2018, 2406594.
  29. Martin, C.R.; Osadchiy, V.; Kalani, A.; Mayer, E.A. The brain-gut-microbiome axis. Cel. Mol. Gastroenterol. Hepat. 2018, 6, 133–148.
  30. Thursby, E.; Juge, N. Introduction to the human gut microbiota. Biochem J. 2017, 16, 1823–1836.
  31. Mayer, E.A.; Tillisch, K.; Gupta, A. Gut/brain axis and the microbiota. J. Clin. Investig. 2015, 125, 926–938.
  32. Miller, A.H.; Raison, C.L. The role of inflammation in depression: From evolutionary imperative to modern treatment target. Nat. Rev. Immunol. 2016, 16, 22–34.
  33. Cryan, J.F.; O’Riordan, K.J.; Cowan, C.S.M.; Sandhu, K.V.; Bastiaanssen, T.F.S.; Boehme, M.; Codagnone, M.G.; Cussotto, S.; Fulling, C.; Golubeva, A.V.; et al. The microbiota-gut-Brain Axis. Physiol. Rev. 2019, 99, 1877–2013.
  34. Galland, L. The gut microbiome and the brain. J. Med. Food. 2014, 17, 1261–1272.
  35. Contestabile, A.; Migani, P.; Poli, A.; Vilani, L.; Bissoli, R.; Cristini, G. Patterns of neurotransmitter function in the optic tectum of teleosts. In Sensory Physiology of Aquatic Lower Vertebrates; Szabó, T., Czéh, G., Eds.; Pergamon: Oxford, UK, 1981; pp. 75–94.
  36. Farzi, A.; Fröhlich, E.E.; Holzer, P. Gut microbiota and the neuroendocrine system. Neurotherapeutics 2018, 15, 5–22.
  37. Yang, D.; Zhao, D.; Ali Shah, S.Z.; Wu, W.; Lai, M.; Zhang, X.; Li, J.; Guan, Z.; Zhao, H.; Li, W.; et al. The role of the gut microbiota in the pathogenesis of Parkinson’s Disease. Front. Neurol. 2019, 10, 1155.
  38. Dutta, S.K.; Verma, S.; Jain, V.; Surapaneni, B.K.; Vinayek, R.; Phillips, L.; Nair, P.P. Parkinson’s Disease: The emerging role of gut dysbiosis, antibiotics, probiotics, and fecal microbiota transplantation. J. Meurogastroenterol. Motil. 2019, 25, 363–376.
  39. Uyar, G.Ö.; Yildiran, H. A nutritional approach to microbiota in Parkinson’s Disease. Biosci. Microbiota Food Health 2019, 38, 115–127.
  40. Ankolekar, C.; Johnson, D.; Pinto, M.S.; Johnson, K.; Labbe, R.; Shetty, K. Inhibitory potential of tea polyphenolics and influence of extraction time against Helicobacter pylori and lack of inhibition of beneficial lactic acid bacteria. J. Med. Food 2011, 14, 1321–1329.
  41. Nakayama, M.; Shigemune, N.; Tsugukuni, T.; Jun, H.; Matsushita, T.; Mekada, Y.; Kurahachi, M.; Miyamoto, T. Mechanism of the combined anti-bacterial effect of green tea extract and NaCl against Staphylococcus aureus and Escherichia coli O157: H7. Food Control. 2012, 25, 225–232.
  42. Xue, R.; Zhang, H.; Pan, J.; Du, Z.; Zhou, W.; Zhang, Z.; Tian, Z.; Zhou, R.; Bai, L. Peripheral dopamine controlled by gut microbes inhibits invariant natural killer T cell-mediated hepatitis. Front. Immunol. 2018, 9, 2398.
  43. Devos, D.; Lebouvier, T.; Lardeux, B.; Biraud, M.; Rouaud, T.; Pouclet, H.; Coron, E.; des Varannes, S.B.; Naveilhan, P.; Nguyen, J.M.; et al. Colonic inflammation in Parkinson’s Disease. Neurobiol. Dis. 2013, 50, 42–48.
  44. Quigley, E.M.; Quera, R. Small intestinal bacterial overgrowth: Roles of antibiotics, prebiotics, and probiotics. Gastroenterology 2006, 130, S78–S90.
  45. Nair, A.; Ramachandran, V.; Joghee, N.; Antony, S.; Ramalingam, G. Gut microbiota dysfunction as reliable non-invasive early diagnostic biomarkers in the pathophysiology of parkinson’s disease. J. Neurogastroenterol. Motil. 2018, 24, 30–42.
  46. Weis, S.; Schwiertz, A.; Unger, M.M.; Becker, A.; Faßbender, K.; Ratering, S.; Kohl, M.; Schnell, S.; Schäfer, K.H.; Egert, M. Effect of Parkinson’s disease and related medications on the composition of the fecal bacterial microbiota. NPJ Parkinsons Dis. 2019, 5, 28.
  47. Singh, R.K.; Chang, H.W.; Yan, D.; Lee, K.M.; Ucmak, D.; Wong, K.; Abrouk, M.; Farahnik, B.; Nakamura, M.; Zhu, T.H.; et al. Influence of diet on the gut microbiome and implications for human health. J. Transl. Med. 2017, 15, 73.
  48. Lerner, A.; Shoenfeld, Y.; Matthias, T. Probiotics: If it does not help it does not do any harm. Really? Microorganisms 2019, 7, 104.
  49. Ilie, O.D.; Ciobica, A.; McKenna, J.; Doroftei, B.; Mavroudis, I. Minireview on the relations between gut microflora and Parkinson’s disease: Further biochemical (oxidative stress), inflammatory, and neurological particularities. Oxid. Med. Cel. Longev. 2020, 2020, 4518023.
  50. Kowalski, K.; Mulak, A. Brain-gut-microbiota axis in Alzheimer’s Disease. J. Neurogastroenterol. Motil. 2019, 25, 48–60.
  51. Carrillo, J.L.M.; Del Campo, J.O.M.; Coronado, O.G.; Gutiérrez, P.T.V.; Cordero, J.F.C.; Juárez, J.V. Adipose tissue and inflammation. In Adipose Tissue; Szablewski, L., Ed.; IntechOpen: London, UK, 2018.
  52. Longo, M.; Zatterale, F.; Naderi, J.; Parrillo, L.; Formisano, P.; Raciti, G.A.; Beguinot, F.; Miele, C. Adipose tissue dysfunction as determinant of obesity-associated metabolic complications. Int. J. Mol. Sci. 2019, 20, 2358.
  53. Cattaneo, A.; Cattane, N.; Galluzzi, S.; Provasi, S.; Lopizzo, N.; Festari, C.; Ferrari, C.; Guerra, U.P.; Paghera, B.; Muscio, C.; et al. Association of brain amyloidosis with pro-inflammatory gut bacterial taxa and peripheral inflammation markers in cognitively impaired elderly. Neurobiol. Aging 2017, 49, 60–68.
  54. Rock, K.D.; Patisaul, H.B. Environmental mechanisms of neurodevelopmental toxicity. Curr. Environ. Health Rep. 2018, 5, 145–157.
  55. Angelucci, F.; Cechova, K.; Amlerova, J.; Hort, J. Antibiotics, gut microbiota, and Alzheimer’s disease. J. Neuroinflamm. 2019, 16, 108.
  56. Askarova, S.; Umbayev, B.; Masoud, A.R.; Kaiyrlykyzy, A.; Safarova, Y.; Tsoy, A.; Olzhayev, F.; Kushugulova, A. The links between the gut microbiome, aging, modern lifestyle and Alzheimer’s Disease. Front. Cell. Infect. Microbiol. 2020, 10, 104.
  57. Vamanu, E.; Gatea, F. Correlations between microbiota bioactivity and bioavailability of functional compounds: A mini-review. Biomedicines 2020, 8, 39.
  58. Blacher, E.; Bashiardes, S.; Shapiro, H.; Rothschild, D.; Mor, U.; Dori-Bachash, M.; Kleimeyer, C.; Moresi, C.; Harnik, Y.; Zur, M.; et al. Potential roles of gut microbiome and metabolites in modulating ALS in mice. Nature 2019, 572, 474–480.
  59. Depommier, C.; Everard, A.; Druart, C.; Plovier, H.; Van Hul, M.; Vieira-Silva, S.; Falony, G.; Raes, J.; Maiter, D.; Delzenne, N.M.; et al. Supplementation with Akkermansia muciniphila in overweight and obese human volunteers: A proof-of-concept exploratory study. Nat. Med. 2019, 25, 1096–1103.
  60. Michaud, A.; Vainik, U.; Garcia-Garcia, I.; Dagher, A. Overlapping neural endophenotypes in addiction and obesity. Front. Endocrinol. 2017, 8, 127.
  61. Volkow, N.D.; Wang, G.J.; Baler, R.D. Reward, dopamine and the control of food intake: Implications for obesity. Trends Cogn. Sci. 2011, 15, 37–46.
  62. Zinöcker, M.K.; Lindseth, I.A. The western diet-microbiome-host interaction and its role in metabolic disease. Nutrients 2018, 10, 365.
  63. Ashrafian, H.; Harling, L.; Darzi, A.; Athanasiou, T. Neurodegenerative disease and obesity: What is the role of weight loss and bariatric interventions? Metab. Brain Dis. 2013, 28, 341–353.
  64. Pugazhenthi, S.; Qin, L.; Reddy, P.H. Common neurodegenerative pathways in obesity, diabetes, and Alzheimer’s disease. Biochim. Biophys. Acta Mol. Basis Dis. 2017, 1863, 1037–1045.
  65. Lerner, A.; McCarty, M.F. The aging bowel dysfunction and elderly vulnerability towards COVID-19 infection. Life 2021, 11, 97.
  66. Luca, M.; Di Mauro, M.; Di Mauro, M.; Luca, A. Gut Microbiota in Alzheimer’s Disease, Depression, and Type 2 Diabetes Mellitus: The Role of Oxidative Stress. Oxid. Med. Cell. Longev. 2019, 2019, 4730539.
  67. Vamanu, E.; Pelinescu, D.; Sarbu, I. Comparative fingerprinting of the human microbiota in diabetes and cardiovascular disease. J. Med. Food. 2016, 19, 1188–1195.
  68. Douglas, B.; Kell, B.D.; Pretorius, E. On the translocation of bacteria and their lipopolysaccharides between blood and peripheral locations in chronic, inflammatory diseases: The central roles of LPS and LPS-induced cell death. Integr. Biol. 2015, 7, 1339–1377.
  69. Loffredo, L.; Ettorre, E.; Zicari, A.M.; Inghilleri, M.; Nocella, C.; Perri, L.; Spalice, A.; Fossati, C.; De Lucia, M.C.; Pigozzi, F.; et al. Neurodegenerative disease study group. oxidative stress and gut-derived lipopolysaccharides in neurodegenerative disease: Role of NOX2. Oxid. Med. Cel. Longev. 2020, 2020, 8630275.
  70. Fiorucci, S.; Carino, A.; Baldoni, M.; Santucci, L.; Costanzi, E.; Graziosi, L.; Distrutti, E.; Bioagioli, M. Bile acid signaling in inflammatory bowel diseases. Dig. Dis. Sci. 2021, 66, 674–693.
  71. Zeng, H.; Umar, S.; Rust, B.; Lazarova, D.; Bordonaro, M. Secondary bile acids and short chain fatty acids in the colon: A focus on colonic microbiome, cell proliferation, inflammation, and cancer. Int. J. Mol. Sci. 2019, 20, 1214.
  72. Agus, A.; Clément, K.; Sokol, H. Gut microbiota-derived metabolites as central regulators in metabolic disorders. Gut 2020.
More
Information
Subjects: Neurosciences
Contributor MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register :
View Times: 608
Revisions: 2 times (View History)
Update Date: 14 Jul 2021
1000/1000