Microbiota modulation counteracts Alzheimer's disease progression influencing neuronal proteolysis and gut hormones plasma levels

Laura Bonfili, Valentina Cecarini, Sara Berardi, Silvia Scarpona, Jan S Suchodolski, Cinzia Nasuti, Dennis Fiorini, Maria Chiara Boarelli, Giacomo Rossi, Anna Maria Eleuteri, Laura Bonfili, Valentina Cecarini, Sara Berardi, Silvia Scarpona, Jan S Suchodolski, Cinzia Nasuti, Dennis Fiorini, Maria Chiara Boarelli, Giacomo Rossi, Anna Maria Eleuteri

Abstract

Gut microbiota has a proven role in regulating multiple neuro-chemical pathways through the highly interconnected gut-brain axis. Oral bacteriotherapy thus has potential in the treatment of central nervous system-related pathologies, such as Alzheimer's disease (AD). Current AD treatments aim to prevent onset, delay progression and ameliorate symptoms. In this work, 3xTg-AD mice in the early stage of AD were treated with SLAB51 probiotic formulation, thereby affecting the composition of gut microbiota and its metabolites. This influenced plasma concentration of inflammatory cytokines and key metabolic hormones considered therapeutic targets in neurodegeneration. Treated mice showed partial restoration of two impaired neuronal proteolytic pathways (the ubiquitin proteasome system and autophagy). Their cognitive decline was decreased compared with controls, due to a reduction in brain damage and reduced accumulation of amyloid beta aggregates. Collectively, our results clearly prove that modulation of the microbiota induces positive effects on neuronal pathways that are able to slow down the progression of Alzheimer's disease.

Conflict of interest statement

The authors declare that they have no competing interests.

Figures

Figure 1
Figure 1
SLAB51 ameliorates behavioral performance and reduces brain damage in AD mice. Novel Object Recognition (NOR) test (first experiment): 15 mice/sub-group were allowed to explore an identical pair of objects, and after 3 hours, they are presented with the familiar object and a novel object. The discrimination scores for 8, 12, 18, and 24-week-old AD mice are reported in panel A. NOR test (second experiment) performed for the first time on treated and untreated 24-week-old mice (groups’ size = 10), panel B. Panel C: Elevated plus maze test. % open arm entries and % time spent in the open arms by untreated and treated AD mice at 24 weeks of age (first experiment, groups’ size = 15). Data points marked with an asterisk are statistically significant compared to their respective non-treated control mice (*p < 0.05). Panel D Immunodetection of FGF9 protein in brain slides of 24-weeks old untreated and treated wt and AD mice (8 animals per group) from the first experiment. Results are reported as number of cells immunoistochemically positive for FGF9 per field ± ES (#statistically significant with respect to the corresponding untreated mice p < 0.05). For each histological section 5 randomly selected field were analyzed at 40xHPFs. Representative images of immunohistochemical staining are reported. Panel E: Brain weights expressed in grams ± ES of both treated and untreated wt and AD mice over time (groups’ size is 8). Panel F: Measurement of the cortex thickness (mm) and ventricular sizes evaluation (mm3) in the brain sections of control and SLAB51 treated AD mice at 8, 12, 18 and 24 weeks of age. Data are reported as mean values ± ES (*statistically significant with respect to 8 weeks-old untreated mice p < 0.05; #statistically significant with respect to the corresponding untreated mice p < 0.05). Consecutive brain slides of treated and untreated 24 weeks-old AD mice are shown.
Figure 2
Figure 2
Microbiota analysis. PCoA plots based on unweighted Unifrac distances and rarefaction curves are presented. The microbiota was analyzed using 16S rRNA gene sequencing. PCoA plots based on unweighted Unifrac distances reveal separation between the microbiota of wt and AD mice at all time points, indicating that the affected mice have different microbiota structure compared to wt mice. No significant differences were found in alpha diversity indices, revealing no difference in species richness. The progression over time from week 8 to week 24 was more pronounced in the AD mice compared to the wt mice. Also, treatment with SLAB51 induced larger changes in AD mice compared to wt mice, but no differences in species richness (t0 = 8 weeks; t1 = 12 weeks; t2 = 18 weeks; t3 = 24 weeks).
Figure 3
Figure 3
Inflammatory cytokines. ELISA of inflammatory cytokines measured in the plasma of 24 week-old wt and AD mice untreated or treated with SLAB51. Analytes concentrations are expressed as mean ± SE. Data points marked with an asterisk are statistically significant compared to their respective untreated mice (*p 

Figure 4

Plasma concentrations of gut hormones.…

Figure 4

Plasma concentrations of gut hormones. Hormones were determined in the plasma of treated…

Figure 4
Plasma concentrations of gut hormones. Hormones were determined in the plasma of treated and control wt and AD mice. Results are expressed as percentage with respect to 8 week-old untreated mice. Data points marked with an asterisk are statistically significant compared to 8 week-old untreated control mice (*p #p < 0.05).

Figure 5

Aβ load. Panel A: Aβ…

Figure 5

Aβ load. Panel A: Aβ 1–40 and Aβ 1–42 levels expressed as pg/ml…

Figure 5
Aβ load. Panel A: Aβ1–40 and Aβ1–42 levels expressed as pg/ml determined by ELISA in the brains of AD mice treated or not with SLAB51 (n = 8). Panel B: Expression levels of amyloid oligomers detected by western blot. The densitometry from five separate blots and a representative immunoblot are reported. Equal protein loading was verified by using an anti-GAPDH antibody. The detection was executed by ECL. Data points marked with an asterisk are statistically significant compared to 8 weeks-old controls (*p < 0.05). Data points marked with hash are statistically significant compared to their respective control mice in the same time point (#p < 0.05). Uncropped gels are reported in Supplemental Figure 1. Panel C: Congo red staining of extra- and intra-cellular amyloid deposits in 24-week-old wt and AD mice administered with water or SLAB51 (groups’ size is 8). Specific Congo red staining was progressively seen in somata and processes of hippocampal Ammon’s horn pyramidal cells (insert), especially in untreated AD mice. Strong extracellular deposits demonstrate the formation of amyloid plaques, also visualized by immunostaining. (Congo red stain, with Meyer’s hematoxylin nuclear counterstain. Coronal sections, Bar = 400 μm; dentate gyrus magnification, Bar = 200 μm; insert, Bar = 50 μm). Data are presented as positive cells/field and are representative of 5 histological section for each brain (n = 8 per sub-group). Data points marked with a hash are statistically significant compared to their respective water-treated mice (p < 0.05). Panel D. Aβ1–42 IHC stain: wt and AD mice administered with water (control) or SLAB51. In both upper (low magnification) and lower (high magnifications) groups of images, immunoreactivity towards Aβ1–42 peptide (Aβ1–42 C-terminus pAb, Millipore) was progressively seen in somata and processes of hippocampal pyramidal cells and cortical neurons of AD untreated mice. A strong extracellular reactivity, associated with Aβ plaque formation, can be observed in both treated and untreated AD mice (IHC stain, with Meyer’s hematoxylin nuclear counterstain. Coronal sections, Bar = 400 μm; dentate gyrus magnification, Bar = 200 μm). The histogram shows the Aβ1–42 positive cells/field. Data represent 5 histological section for each brain (n = 8). Data points marked with a hash are statistically significant compared to their respective water-treated mice (p < 0.05).

Figure 6

Effect of SLAB51 on proteasomal…

Figure 6

Effect of SLAB51 on proteasomal activity. Proteasome activity in SLAB51 treated and untreated…

Figure 6
Effect of SLAB51 on proteasomal activity. Proteasome activity in SLAB51 treated and untreated wt (left) and AD (right) mice. The ChT-L, T-L, PGPH and BrAAP activities of the 20S proteasome and the ChT-L activity of the 26S proteasome were measured in brain homogenates as described in the Methods section. Results are expressed as fluorescence units (U. F.). Data points marked with an asterisk are statistically significant compared to untreated 8-week-old mice (*p 

Figure 7

Effect of SLAB51 on proteasomal…

Figure 7

Effect of SLAB51 on proteasomal substrates. Detection of the levels of ubiquitinated proteins…

Figure 7
Effect of SLAB51 on proteasomal substrates. Detection of the levels of ubiquitinated proteins (panel A), p27 (panel B) and p53 (panel C) in SLAB51 treated and untreated wt and AD mice. The densitometric analyses obtained from five separate blots and representative immunoblots are shown. Equal protein loading was verified by using an anti-GAPDH antibody. The detection was executed by an ECL western blotting analysis system. Data points marked with an asterisk are statistically significant compared to 8-week-old control mice (*p #p < 0.05). Uncropped gels are reported in Supplemental Figure 1.

Figure 8

TUNEL detection of apoptotic neurons…

Figure 8

TUNEL detection of apoptotic neurons in hippocampal area of SLAB51 treated and untreated…

Figure 8
TUNEL detection of apoptotic neurons in hippocampal area of SLAB51 treated and untreated AD and wt mice. Apoptotic cells are characterized by black-brownish nuclear stain, as shown in the representative images. (TUNEL (DeadEnd, Promega®) reaction, with Meyer’s hematoxylin nuclear counterstain. Bar = 200 μm.). The histogram shows the TUNEL positive cells/field. Data are representative of 5 histological section for each brain (n = 8 per sub-group). Data points marked with a hash are statistically significant compared to their respective water-treated mice (p 

Figure 9

Autophagic markers. Panel A: Cathepsin…

Figure 9

Autophagic markers. Panel A: Cathepsin B and cathepsin L activity in SLAB51 treated…

Figure 9
Autophagic markers. Panel A: Cathepsin B and cathepsin L activity in SLAB51 treated and untreated wt and AD mice. Results are expressed as fluorescence units. Data points marked with an asterisk are statistically significant compared to their respective untreated control mice (*p #p < 0.05) Original membranes strips are reported in Supplemental Figure 1.
All figures (9)
Similar articles
Cited by
References
    1. Cecarini V, et al. Crosstalk between the ubiquitin-proteasome system and autophagy in a human cellular model of Alzheimer’s disease. Biochimica et biophysica acta. 2012;1822:1741–1751. doi: 10.1016/j.bbadis.2012.07.015. - DOI - PubMed
    1. Zheng Q, Li J, Wang X. Interplay between the ubiquitin-proteasome system and autophagy in proteinopathies. International journal of physiology, pathophysiology and pharmacology. 2009;1:127–142. - PMC - PubMed
    1. Nilsson P, et al. Abeta secretion and plaque formation depend on autophagy. Cell reports. 2013;5:61–69. doi: 10.1016/j.celrep.2013.08.042. - DOI - PubMed
    1. van Tijn P, et al. Mutant ubiquitin decreases amyloid beta plaque formation in a transgenic mouse model of Alzheimer’s disease. Neurochemistry international. 2012;61:739–748. doi: 10.1016/j.neuint.2012.07.007. - DOI - PubMed
    1. Gomes S, et al. Protective effect of leptin and ghrelin against toxicity induced by amyloid-beta oligomers in a hypothalamic cell line. Journal of neuroendocrinology. 2014;26:176–185. doi: 10.1111/jne.12138. - DOI - PubMed
Show all 97 references
Publication types
MeSH terms
Related information
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM

NCBI Literature Resources

MeSH PMC Bookshelf Disclaimer

The PubMed wordmark and PubMed logo are registered trademarks of the U.S. Department of Health and Human Services (HHS). Unauthorized use of these marks is strictly prohibited.

Follow NCBI
Figure 4
Figure 4
Plasma concentrations of gut hormones. Hormones were determined in the plasma of treated and control wt and AD mice. Results are expressed as percentage with respect to 8 week-old untreated mice. Data points marked with an asterisk are statistically significant compared to 8 week-old untreated control mice (*p #p < 0.05).
Figure 5
Figure 5
Aβ load. Panel A: Aβ1–40 and Aβ1–42 levels expressed as pg/ml determined by ELISA in the brains of AD mice treated or not with SLAB51 (n = 8). Panel B: Expression levels of amyloid oligomers detected by western blot. The densitometry from five separate blots and a representative immunoblot are reported. Equal protein loading was verified by using an anti-GAPDH antibody. The detection was executed by ECL. Data points marked with an asterisk are statistically significant compared to 8 weeks-old controls (*p < 0.05). Data points marked with hash are statistically significant compared to their respective control mice in the same time point (#p < 0.05). Uncropped gels are reported in Supplemental Figure 1. Panel C: Congo red staining of extra- and intra-cellular amyloid deposits in 24-week-old wt and AD mice administered with water or SLAB51 (groups’ size is 8). Specific Congo red staining was progressively seen in somata and processes of hippocampal Ammon’s horn pyramidal cells (insert), especially in untreated AD mice. Strong extracellular deposits demonstrate the formation of amyloid plaques, also visualized by immunostaining. (Congo red stain, with Meyer’s hematoxylin nuclear counterstain. Coronal sections, Bar = 400 μm; dentate gyrus magnification, Bar = 200 μm; insert, Bar = 50 μm). Data are presented as positive cells/field and are representative of 5 histological section for each brain (n = 8 per sub-group). Data points marked with a hash are statistically significant compared to their respective water-treated mice (p < 0.05). Panel D. Aβ1–42 IHC stain: wt and AD mice administered with water (control) or SLAB51. In both upper (low magnification) and lower (high magnifications) groups of images, immunoreactivity towards Aβ1–42 peptide (Aβ1–42 C-terminus pAb, Millipore) was progressively seen in somata and processes of hippocampal pyramidal cells and cortical neurons of AD untreated mice. A strong extracellular reactivity, associated with Aβ plaque formation, can be observed in both treated and untreated AD mice (IHC stain, with Meyer’s hematoxylin nuclear counterstain. Coronal sections, Bar = 400 μm; dentate gyrus magnification, Bar = 200 μm). The histogram shows the Aβ1–42 positive cells/field. Data represent 5 histological section for each brain (n = 8). Data points marked with a hash are statistically significant compared to their respective water-treated mice (p < 0.05).
Figure 6
Figure 6
Effect of SLAB51 on proteasomal activity. Proteasome activity in SLAB51 treated and untreated wt (left) and AD (right) mice. The ChT-L, T-L, PGPH and BrAAP activities of the 20S proteasome and the ChT-L activity of the 26S proteasome were measured in brain homogenates as described in the Methods section. Results are expressed as fluorescence units (U. F.). Data points marked with an asterisk are statistically significant compared to untreated 8-week-old mice (*p 

Figure 7

Effect of SLAB51 on proteasomal…

Figure 7

Effect of SLAB51 on proteasomal substrates. Detection of the levels of ubiquitinated proteins…

Figure 7
Effect of SLAB51 on proteasomal substrates. Detection of the levels of ubiquitinated proteins (panel A), p27 (panel B) and p53 (panel C) in SLAB51 treated and untreated wt and AD mice. The densitometric analyses obtained from five separate blots and representative immunoblots are shown. Equal protein loading was verified by using an anti-GAPDH antibody. The detection was executed by an ECL western blotting analysis system. Data points marked with an asterisk are statistically significant compared to 8-week-old control mice (*p #p < 0.05). Uncropped gels are reported in Supplemental Figure 1.

Figure 8

TUNEL detection of apoptotic neurons…

Figure 8

TUNEL detection of apoptotic neurons in hippocampal area of SLAB51 treated and untreated…

Figure 8
TUNEL detection of apoptotic neurons in hippocampal area of SLAB51 treated and untreated AD and wt mice. Apoptotic cells are characterized by black-brownish nuclear stain, as shown in the representative images. (TUNEL (DeadEnd, Promega®) reaction, with Meyer’s hematoxylin nuclear counterstain. Bar = 200 μm.). The histogram shows the TUNEL positive cells/field. Data are representative of 5 histological section for each brain (n = 8 per sub-group). Data points marked with a hash are statistically significant compared to their respective water-treated mice (p 

Figure 9

Autophagic markers. Panel A: Cathepsin…

Figure 9

Autophagic markers. Panel A: Cathepsin B and cathepsin L activity in SLAB51 treated…

Figure 9
Autophagic markers. Panel A: Cathepsin B and cathepsin L activity in SLAB51 treated and untreated wt and AD mice. Results are expressed as fluorescence units. Data points marked with an asterisk are statistically significant compared to their respective untreated control mice (*p #p < 0.05) Original membranes strips are reported in Supplemental Figure 1.
All figures (9)
Similar articles
Cited by
References
    1. Cecarini V, et al. Crosstalk between the ubiquitin-proteasome system and autophagy in a human cellular model of Alzheimer’s disease. Biochimica et biophysica acta. 2012;1822:1741–1751. doi: 10.1016/j.bbadis.2012.07.015. - DOI - PubMed
    1. Zheng Q, Li J, Wang X. Interplay between the ubiquitin-proteasome system and autophagy in proteinopathies. International journal of physiology, pathophysiology and pharmacology. 2009;1:127–142. - PMC - PubMed
    1. Nilsson P, et al. Abeta secretion and plaque formation depend on autophagy. Cell reports. 2013;5:61–69. doi: 10.1016/j.celrep.2013.08.042. - DOI - PubMed
    1. van Tijn P, et al. Mutant ubiquitin decreases amyloid beta plaque formation in a transgenic mouse model of Alzheimer’s disease. Neurochemistry international. 2012;61:739–748. doi: 10.1016/j.neuint.2012.07.007. - DOI - PubMed
    1. Gomes S, et al. Protective effect of leptin and ghrelin against toxicity induced by amyloid-beta oligomers in a hypothalamic cell line. Journal of neuroendocrinology. 2014;26:176–185. doi: 10.1111/jne.12138. - DOI - PubMed
Show all 97 references
Publication types
MeSH terms
Related information
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM
Figure 7
Figure 7
Effect of SLAB51 on proteasomal substrates. Detection of the levels of ubiquitinated proteins (panel A), p27 (panel B) and p53 (panel C) in SLAB51 treated and untreated wt and AD mice. The densitometric analyses obtained from five separate blots and representative immunoblots are shown. Equal protein loading was verified by using an anti-GAPDH antibody. The detection was executed by an ECL western blotting analysis system. Data points marked with an asterisk are statistically significant compared to 8-week-old control mice (*p #p < 0.05). Uncropped gels are reported in Supplemental Figure 1.
Figure 8
Figure 8
TUNEL detection of apoptotic neurons in hippocampal area of SLAB51 treated and untreated AD and wt mice. Apoptotic cells are characterized by black-brownish nuclear stain, as shown in the representative images. (TUNEL (DeadEnd, Promega®) reaction, with Meyer’s hematoxylin nuclear counterstain. Bar = 200 μm.). The histogram shows the TUNEL positive cells/field. Data are representative of 5 histological section for each brain (n = 8 per sub-group). Data points marked with a hash are statistically significant compared to their respective water-treated mice (p 

Figure 9

Autophagic markers. Panel A: Cathepsin…

Figure 9

Autophagic markers. Panel A: Cathepsin B and cathepsin L activity in SLAB51 treated…

Figure 9
Autophagic markers. Panel A: Cathepsin B and cathepsin L activity in SLAB51 treated and untreated wt and AD mice. Results are expressed as fluorescence units. Data points marked with an asterisk are statistically significant compared to their respective untreated control mice (*p #p < 0.05) Original membranes strips are reported in Supplemental Figure 1.
All figures (9)
Figure 9
Figure 9
Autophagic markers. Panel A: Cathepsin B and cathepsin L activity in SLAB51 treated and untreated wt and AD mice. Results are expressed as fluorescence units. Data points marked with an asterisk are statistically significant compared to their respective untreated control mice (*p #p < 0.05) Original membranes strips are reported in Supplemental Figure 1.

References

    1. Cecarini V, et al. Crosstalk between the ubiquitin-proteasome system and autophagy in a human cellular model of Alzheimer’s disease. Biochimica et biophysica acta. 2012;1822:1741–1751. doi: 10.1016/j.bbadis.2012.07.015.
    1. Zheng Q, Li J, Wang X. Interplay between the ubiquitin-proteasome system and autophagy in proteinopathies. International journal of physiology, pathophysiology and pharmacology. 2009;1:127–142.
    1. Nilsson P, et al. Abeta secretion and plaque formation depend on autophagy. Cell reports. 2013;5:61–69. doi: 10.1016/j.celrep.2013.08.042.
    1. van Tijn P, et al. Mutant ubiquitin decreases amyloid beta plaque formation in a transgenic mouse model of Alzheimer’s disease. Neurochemistry international. 2012;61:739–748. doi: 10.1016/j.neuint.2012.07.007.
    1. Gomes S, et al. Protective effect of leptin and ghrelin against toxicity induced by amyloid-beta oligomers in a hypothalamic cell line. Journal of neuroendocrinology. 2014;26:176–185. doi: 10.1111/jne.12138.
    1. Folch J, et al. The role of leptin in the sporadic form of Alzheimer’s disease. Interactions with the adipokines amylin, ghrelin and the pituitary hormone prolactin. Life sciences. 2015
    1. Theodoropoulou A, Metallinos IC, Psyrogiannis A, Vagenakis GA, Kyriazopoulou V. Ghrelin and leptin secretion in patients with moderate Alzheimer’s disease. The journal of nutrition, health & aging. 2012;16:472–477. doi: 10.1007/s12603-012-0058-4.
    1. Stoyanova. Ghrelin: A link between ageing, metabolism and neurodegenerative disorders. Neurobiology of Disease, 1–12 (2014).
    1. Moon M, et al. Ghrelin ameliorates cognitive dysfunction and neurodegeneration in intrahippocampal amyloid-beta1-42 oligomer-injected mice. Journal of Alzheimer’s disease: JAD. 2011;23:147–159.
    1. Niedowicz DM, et al. Leptin regulates amyloid beta production via the gamma-secretase complex. Biochimica et biophysica acta. 2013;1832:439–444. doi: 10.1016/j.bbadis.2012.12.009.
    1. Fewlass DC, et al. Obesity-related leptin regulates Alzheimer’s Abeta. FASEB journal: official publication of the Federation of American Societies for Experimental Biology. 2004;18:1870–1878. doi: 10.1096/fj.04-2572com.
    1. Greco SJ, et al. Leptin reduces pathology and improves memory in a transgenic mouse model of Alzheimer’s disease. Journal of Alzheimer’s disease: JAD. 2010;19:1155–1167. doi: 10.3233/JAD-2010-1308.
    1. Rigamonti AE, et al. Plasma ghrelin concentrations in elderly subjects: comparison with anorexic and obese patients. The Journal of endocrinology. 2002;175:R1–5. doi: 10.1677/joe.0.175R001.
    1. Perry T, et al. Glucagon-like peptide-1 decreases endogenous amyloid-beta peptide (Abeta) levels and protects hippocampal neurons from death induced by Abeta and iron. Journal of neuroscience research. 2003;72:603–612. doi: 10.1002/jnr.10611.
    1. Abbas T, Faivre E, Holscher C. Impairment of synaptic plasticity and memory formation in GLP-1 receptor KO mice: Interaction between type 2 diabetes and Alzheimer’s disease. Behavioural brain research. 2009;205:265–271. doi: 10.1016/j.bbr.2009.06.035.
    1. Gault VA, Holscher C. Protease-resistant glucose-dependent insulinotropic polypeptide agonists facilitate hippocampal LTP and reverse the impairment of LTP induced by beta-amyloid. Journal of neurophysiology. 2008;99:1590–1595. doi: 10.1152/jn.01161.2007.
    1. Faivre E, Holscher C. Neuroprotective effects of D-Ala(2)GIP on Alzheimer’s disease biomarkers in an APP/PS1 mouse model. Alzheimer’s research & therapy. 2013;5:20. doi: 10.1186/alzrt174.
    1. Bhattacharjee S, Lukiw WJ. Alzheimer’s disease and the microbiome. Frontiers in cellular neuroscience. 2013;7:153. doi: 10.3389/fncel.2013.00153.
    1. Wang Y, Kasper LH. The role of microbiome in central nervous system disorders. Brain, behavior, and immunity. 2014;38:1–12. doi: 10.1016/j.bbi.2013.12.015.
    1. Kang SS, et al. Diet and exercise orthogonally alter the gut microbiome and reveal independent associations with anxiety and cognition. Molecular neurodegeneration. 2014;9:36. doi: 10.1186/1750-1326-9-36.
    1. Tang, M. L. et al. Administration of a probiotic with peanut oral immunotherapy: A randomized trial. The Journal of allergy and clinical immunology135, 737–744 e738, doi:10.1016/j.jaci.2014.11.034 (2015).
    1. Penha Filho, R. A. et al. Immunomodulatory activity and control of Salmonella Enteritidis colonization in the intestinal tract of chickens by Lactobacillus based probiotic. Veterinary immunology and immunopathology, doi:10.1016/j.vetimm.2015.06.006 (2015).
    1. Fedorak, R. N. et al. The probiotic VSL#3 has anti-inflammatory effects and could reduce endoscopic recurrence after surgery for Crohn’s disease. Clinical gastroenterology and hepatology: the official clinical practice journal of the American Gastroenterological Association13, 928–935 e922, doi:10.1016/j.cgh.2014.10.031 (2015).
    1. Rossi G, et al. Comparison of microbiological, histological, and immunomodulatory parameters in response to treatment with either combination therapy with prednisone and metronidazole or probiotic VSL#3 strains in dogs with idiopathic inflammatory bowel disease. PloS one. 2014;9:e94699. doi: 10.1371/journal.pone.0094699.
    1. Talero E, et al. Inhibition of chronic ulcerative colitis-associated adenocarcinoma development in mice by VSL#3. Inflammatory bowel diseases. 2015;21:1027–1037. doi: 10.1097/MIB.0000000000000346.
    1. Duncan SH, Flint HJ. Probiotics and prebiotics and health in ageing populations. Maturitas. 2013;75:44–50. doi: 10.1016/j.maturitas.2013.02.004.
    1. Saulnier DM, et al. The intestinal microbiome, probiotics and prebiotics in neurogastroenterology. Gut microbes. 2013;4:17–27. doi: 10.4161/gmic.22973.
    1. von Geldern G, Mowry EM. The influence of nutritional factors on the prognosis of multiple sclerosis. Nature reviews. Neurology. 2012;8:678–689. doi: 10.1038/nrneurol.2012.194.
    1. Camfield DA, Owen L, Scholey AB, Pipingas A, Stough C. Dairy constituents and neurocognitive health in ageing. The British journal of nutrition. 2011;106:159–174. doi: 10.1017/S0007114511000158.
    1. Douglas-Escobar M, Elliott E, Neu J. Effect of intestinal microbial ecology on the developing brain. JAMA pediatrics. 2013;167:374–379. doi: 10.1001/jamapediatrics.2013.497.
    1. Hsiao EY, et al. Microbiota modulate behavioral and physiological abnormalities associated with neurodevelopmental disorders. Cell. 2013;155:1451–1463. doi: 10.1016/j.cell.2013.11.024.
    1. Davari S, Talaei SA, Alaei H, Salami M. Probiotics treatment improves diabetes-induced impairment of synaptic activity and cognitive function: behavioral and electrophysiological proofs for microbiome-gut-brain axis. Neuroscience. 2013;240:287–296. doi: 10.1016/j.neuroscience.2013.02.055.
    1. Distrutti E, et al. Modulation of intestinal microbiota by the probiotic VSL#3 resets brain gene expression and ameliorates the age-related deficit in LTP. PloS one. 2014;9:e106503. doi: 10.1371/journal.pone.0106503.
    1. Vinolo MA, Rodrigues HG, Nachbar RT, Curi R. Regulation of inflammation by short chain fatty acids. Nutrients. 2011;3:858–876. doi: 10.3390/nu3100858.
    1. Oleskin AV, Shenderov BA. Neuromodulatory effects and targets of the SCFAs and gasotransmitters produced by the human symbiotic microbiota. Microbial ecology in health and disease. 2016;27:30971.
    1. Orlowski M, Michaud C. Pituitary multicatalytic proteinase complex. Specificity of components and aspects of proteolytic activity. Biochemistry. 1989;28:9270–9278.
    1. Pfleiderer G, Celliers PG. [Isolation of an Aminopeptidase from Kidney Particles] Biochemische Zeitschrift. 1963;339:186–189.
    1. Oddo S, et al. Triple-transgenic model of Alzheimer’s disease with plaques and tangles: intracellular Abeta and synaptic dysfunction. Neuron. 2003;39:409–421. doi: 10.1016/S0896-6273(03)00434-3.
    1. Crawford JD, Terry ME, Rourke GM. Simplification of drug dosage calculation by application of the surface area principle. Pediatrics. 1950;5:783–790.
    1. Nasuti C, et al. Dopaminergic system modulation, behavioral changes, and oxidative stress after neonatal administration of pyrethroids. Toxicology. 2007;229:194–205. doi: 10.1016/j.tox.2006.10.015.
    1. Leger M, et al. Object recognition test in mice. Nature protocols. 2013;8:2531–2537. doi: 10.1038/nprot.2013.155.
    1. Nasuti C, et al. Neonatal exposure to permethrin pesticide causes lifelong fear and spatial learning deficits and alters hippocampal morphology of synapses. Journal of neurodevelopmental disorders. 2014;6:7. doi: 10.1186/1866-1955-6-7.
    1. Kim HY, et al. Taurine in drinking water recovers learning and memory in the adult APP/PS1 mouse model of Alzheimer’s disease. Scientific reports. 2014;4:7467. doi: 10.1038/srep07467.
    1. Lister RG. The use of a plus-maze to measure anxiety in the mouse. Psychopharmacology. 1987;92:180–185.
    1. Nasuti C, et al. Effects of early life permethrin exposure on spatial working memory and on monoamine levels in different brain areas of pre-senescent rats. Toxicology. 2013;303:162–168. doi: 10.1016/j.tox.2012.09.016.
    1. Bell ET, et al. Faecal microbiota of cats with insulin-treated diabetes mellitus. PloS one. 2014;9:e108729. doi: 10.1371/journal.pone.0108729.
    1. McDonald D, et al. An improved Greengenes taxonomy with explicit ranks for ecological and evolutionary analyses of bacteria and archaea. The ISME journal. 2012;6:610–618. doi: 10.1038/ismej.2011.139.
    1. Fiorini D, Boarelli MC, Gabbianelli R, Ballini R, Pacetti D. A quantitative headspace-solid-phase microextraction-gas chromatography-flame ionization detector method to analyze short chain free fatty acids in rat feces. Analytical biochemistry. 2016;508:12–14. doi: 10.1016/j.ab.2016.05.023.
    1. Paxinos, G. & Franklin, K. B. J. The mouse brain in stereotaxic coordinates (Academic Press, 2001).
    1. Liu P, et al. Characterization of a Novel Mouse Model of Alzheimer’s Disease–Amyloid Pathology and Unique beta-Amyloid Oligomer Profile. PloS one. 2015;10:e0126317. doi: 10.1371/journal.pone.0126317.
    1. Bradford MM. A rapid and sensitive method for the quantitation of microgram quantities of protein utilizing the principle of protein-dye binding. Analytical biochemistry. 1976;72:248–254. doi: 10.1016/0003-2697(76)90527-3.
    1. Eleuteri AM, et al. Isolation and characterization of bovine thymus multicatalytic proteinase complex. Protein expression and purification. 2000;18:160–168. doi: 10.1006/prep.1999.1187.
    1. Tchoupe JR, Moreau T, Gauthier F, Bieth JG. Photometric or fluorometric assay of cathepsin B, L and H and papain using substrates with an aminotrifluoromethylcoumarin leaving group. Biochimica et biophysica acta. 1991;1076:149–151. doi: 10.1016/0167-4838(91)90232-O.
    1. Schneider CA, Rasband WS, Eliceiri KW. NIH Image to ImageJ: 25 years of image analysis. Nature methods. 2012;9:671–675. doi: 10.1038/nmeth.2089.
    1. Marchini C, Angeletti M, Eleuteri AM, Fedeli A, Fioretti E. Aspirin modulates LPS-induced nitric oxide release in rat glial cells. Neuroscience letters. 2005;381:86–91. doi: 10.1016/j.neulet.2005.02.002.
    1. Evans SJ, et al. Dysregulation of the fibroblast growth factor system in major depression. Proceedings of the National Academy of Sciences of the United States of America. 2004;101:15506–15511. doi: 10.1073/pnas.0406788101.
    1. Bernard R, et al. Altered expression of glutamate signaling, growth factor, and glia genes in the locus coeruleus of patients with major depression. Molecular psychiatry. 2011;16:634–646. doi: 10.1038/mp.2010.44.
    1. Gaughran F, Payne J, Sedgwick PM, Cotter D, Berry M. Hippocampal FGF-2 and FGFR1 mRNA expression in major depression, schizophrenia and bipolar disorder. Brain research bulletin. 2006;70:221–227. doi: 10.1016/j.brainresbull.2006.04.008.
    1. Allen-Vercoe E, et al. Anaerostipes hadrus comb. nov., a dominant species within the human colonic microbiota; reclassification of Eubacterium hadrum Moore et al. 1976. Anaerobe. 2012;18:523–529. doi: 10.1016/j.anaerobe.2012.09.002.
    1. Sanchez-Ramos J, et al. Granulocyte colony stimulating factor decreases brain amyloid burden and reverses cognitive impairment in Alzheimer’s mice. Neuroscience. 2009;163:55–72. doi: 10.1016/j.neuroscience.2009.05.071.
    1. McKinnon C, Tabrizi SJ. The ubiquitin-proteasome system in neurodegeneration. Antioxidants & redox signaling. 2014;21:2302–2321. doi: 10.1089/ars.2013.5802.
    1. Hook G, Yu J, Toneff T, Kindy M, Hook V. Brain pyroglutamate amyloid-beta is produced by cathepsin B and is reduced by the cysteine protease inhibitor E64d, representing a potential Alzheimer’s disease therapeutic. Journal of Alzheimer’s disease: JAD. 2014;41:129–149.
    1. Klein DM, Felsenstein KM, Brenneman DE. Cathepsins B and L differentially regulate amyloid precursor protein processing. The Journal of pharmacology and experimental therapeutics. 2009;328:813–821. doi: 10.1124/jpet.108.147082.
    1. Kang R, Zeh HJ, Lotze MT, Tang D. The Beclin 1 network regulates autophagy and apoptosis. Cell death and differentiation. 2011;18:571–580. doi: 10.1038/cdd.2010.191.
    1. Klionsky DJ, et al. Guidelines for the use and interpretation of assays for monitoring autophagy. Autophagy. 2012;8:445–544. doi: 10.4161/auto.19496.
    1. Bjorkoy G, et al. Monitoring autophagic degradation of p62/SQSTM1. Methods in enzymology. 2009;452:181–197. doi: 10.1016/S0076-6879(08)03612-4.
    1. Sultana R, Perluigi M, Butterfield DA. Oxidatively modified proteins in Alzheimer’s disease (AD), mild cognitive impairment and animal models of AD: role of Abeta in pathogenesis. Acta neuropathologica. 2009;118:131–150. doi: 10.1007/s00401-009-0517-0.
    1. Takashima A. A-beta, tau, and dementia. Journal of Alzheimer’s disease: JAD. 2009;17:729–736. doi: 10.3233/JAD-2009-1090.
    1. Bercik P, Collins SM, Verdu EF. Microbes and the gut-brain axis. Neurogastroenterology and motility: the official journal of the European Gastrointestinal Motility Society. 2012;24:405–413. doi: 10.1111/j.1365-2982.2012.01906.x.
    1. Distrutti E, Cipriani S, Mencarelli A, Renga B, Fiorucci S. Probiotics VSL#3 protect against development of visceral pain in murine model of irritable bowel syndrome. PloS one. 2013;8:e63893. doi: 10.1371/journal.pone.0063893.
    1. Khokhlova EV, et al. Anti-inflammatory properties of intestinal Bifidobacterium strains isolated from healthy infants. Microbiology and immunology. 2012;56:27–39. doi: 10.1111/j.1348-0421.2011.00398.x.
    1. Okada Y, et al. Anti-inflammatory effects of the genus Bifidobacterium on macrophages by modification of phospho-I kappaB and SOCS gene expression. International journal of experimental pathology. 2009;90:131–140. doi: 10.1111/j.1365-2613.2008.00632.x.
    1. Hamza E, Kittl S, Kuhnert P. Temporal induction of pro-inflammatory and regulatory cytokines in human peripheral blood mononuclear cells by Campylobacter jejuni and Campylobacter coli. PloS one. 2017;12:e0171350. doi: 10.1371/journal.pone.0171350.
    1. Barjesteh N, et al. Induction of chicken cytokine responses in vivo and in vitro by lipooligosaccharide of Campylobacter jejuni HS:10. Veterinary microbiology. 2013;164:122–130. doi: 10.1016/j.vetmic.2013.02.002.
    1. Barrett E, Ross RP, O’Toole PW, Fitzgerald GF, Stanton C. gamma-Aminobutyric acid production by culturable bacteria from the human intestine. Journal of applied microbiology. 2012;113:411–417. doi: 10.1111/j.1365-2672.2012.05344.x.
    1. Lyte M, Varcoe JJ, Bailey MT. Anxiogenic effect of subclinical bacterial infection in mice in the absence of overt immune activation. Physiology & behavior. 1998;65:63–68. doi: 10.1016/S0031-9384(98)00145-0.
    1. Kountouras J, et al. Eradication of Helicobacter pylori may be beneficial in the management of Alzheimer’s disease. Journal of neurology. 2009;256:758–767. doi: 10.1007/s00415-009-5011-z.
    1. Irving AJ, Harvey J. Leptin regulation of hippocampal synaptic function in health and disease. Philosophical transactions of the Royal Society of London. Series B, Biological sciences. 2014;369:20130155. doi: 10.1098/rstb.2013.0155.
    1. Hebda-Bauer EK, et al. 3xTg-AD mice exhibit an activated central stress axis during early-stage pathology. Journal of Alzheimer’s disease: JAD. 2013;33:407–422.
    1. Malcher-Lopes R, et al. Opposing crosstalk between leptin and glucocorticoids rapidly modulates synaptic excitation via endocannabinoid release. The Journal of neuroscience: the official journal of the Society for Neuroscience. 2006;26:6643–6650. doi: 10.1523/JNEUROSCI.5126-05.2006.
    1. Holscher C. The incretin hormones glucagonlike peptide 1 and glucose-dependent insulinotropic polypeptide are neuroprotective in mouse models of Alzheimer’s disease. Alzheimer’s & dementia: the journal of the Alzheimer’s Association. 2014;10:S47–54. doi: 10.1016/j.jalz.2013.12.009.
    1. Cox MA, et al. Short-chain fatty acids act as antiinflammatory mediators by regulating prostaglandin E(2) and cytokines. World journal of gastroenterology. 2009;15:5549–5557. doi: 10.3748/wjg.15.5549.
    1. Russo, R. et al. Gut-brain axis: Role of lipids in the regulation of inflammation, pain and CNS diseases. Current medicinal chemistry, doi:10.2174/0929867324666170216113756 (2017).
    1. Lei E, Vacy K, Boon WC. Fatty acids and their therapeutic potential in neurological disorders. Neurochemistry international. 2016;95:75–84. doi: 10.1016/j.neuint.2016.02.014.
    1. Ricobaraza A, Cuadrado-Tejedor M, Marco S, Perez-Otano I, Garcia-Osta A. Phenylbutyrate rescues dendritic spine loss associated with memory deficits in a mouse model of Alzheimer disease. Hippocampus. 2012;22:1040–1050. doi: 10.1002/hipo.20883.
    1. Frost G, et al. The short-chain fatty acid acetate reduces appetite via a central homeostatic mechanism. Nature communications. 2014;5:3611. doi: 10.1038/ncomms4611.
    1. Ohsawa K, Uchida N, Ohki K, Nakamura Y, Yokogoshi H. Lactobacillus helveticus-fermented milk improves learning and memory in mice. Nutritional neuroscience. 2015;18:232–240. doi: 10.1179/1476830514Y.0000000122.
    1. Keller JN, Hanni KB, Markesbery WR. Impaired proteasome function in Alzheimer’s disease. Journal of neurochemistry. 2000;75:436–439. doi: 10.1046/j.1471-4159.2000.0750436.x.
    1. Kitamura Y, et al. Changes of p53 in the brains of patients with Alzheimer’s disease. Biochemical and biophysical research communications. 1997;232:418–421. doi: 10.1006/bbrc.1997.6301.
    1. Ohyagi Y, et al. Intracellular Abeta42 activates p53 promoter: a pathway to neurodegeneration in Alzheimer’s disease. FASEB journal: official publication of the Federation of American Societies for Experimental Biology. 2005;19:255–257.
    1. de la Monte SM, Sohn YK, Wands JR. Correlates of p53- and Fas (CD95)-mediated apoptosis in Alzheimer’s disease. Journal of the neurological sciences. 1997;152:73–83. doi: 10.1016/S0022-510X(97)00131-7.
    1. Hooper C, et al. p53 is upregulated in Alzheimer’s disease and induces tau phosphorylation in HEK293a cells. Neuroscience letters. 2007;418:34–37. doi: 10.1016/j.neulet.2007.03.026.
    1. Cecarini V, et al. Effects of Ghrelin on the Proteolytic Pathways of Alzheimer’s Disease Neuronal Cells. Molecular neurobiology. 2015
    1. Ulloa F, Marti E. Wnt won the war: antagonistic role of Wnt over Shh controls dorso-ventral patterning of the vertebrate neural tube. Developmental dynamics: an official publication of the American Association of Anatomists. 2010;239:69–76.
    1. Aurbach EL, et al. Fibroblast growth factor 9 is a novel modulator of negative affect. Proceedings of the National Academy of Sciences of the United States of America. 2015;112:11953–11958. doi: 10.1073/pnas.1510456112.
    1. Nakamura S, et al. Fibroblast growth factor (FGF)-9 immunoreactivity in senile plaques. Brain research. 1998;814:222–225. doi: 10.1016/S0006-8993(98)01042-7.
    1. Diaz Heijtz R, et al. Normal gut microbiota modulates brain development and behavior. Proceedings of the National Academy of Sciences of the United States of America. 2011;108:3047–3052. doi: 10.1073/pnas.1010529108.

Source: PubMed

3
Sottoscrivi